Blood Safety and SURVEILLANCE PDF
Blood Safety and SURVEILLANCE PDF
Blood Safety and SURVEILLANCE PDF
SAFETY
and
SURVEILLANCE
edited by
Jeanne V. Linden
Wadsworth Center
New York State Department of Health
Albany, New York
Celso Bianco
New York Blood Center
New York, New York
Headquarters
Marcel Dekker, Inc.
270 Madison Avenue, New York, NY 10016
tel: 212-696-9000; fax: 212-685-4540
The publisher offers discounts on this book when ordered in bulk quantities. For more
information, write to Special Sales/Professional Marketing at the headquarters address
above.
Neither this book nor any part may be reproduced or transmitted in any form or by any
means, electronic or mechanical, including photocopying, microfilming, and recording, or
by any information storage and retrieval system, without permission in writing from the
publisher.
Foreword
Such was the claim of the Chinese in the year 1000 B.C.! Since that time, the
importance of blood has largely moved from the spiritual to the medicinal arena.
Over the centuries, belief in the therapeutic benefits of blood has evolved in a
dramatic manner. At one time in the Roman empire, gladiators drank the blood
of their fallen opponents to reinvigorate themselves. Elsewhere, others sought
rebirth from being showered with the blood of a sacrificed bull.
The first blood transfusion is credited to Richard Lower, who transfused the
blood of one dog into another in 1665 (1). In July 1667, Jean Dennis, a young
French physician, performed the first known human transfusion using lamb blood,
preceding Lowers similar attempts by only a few months:
The project of causing the blood of an healthy animal to pass into the veins
of one diseased, having been conceived about ten years ago, in the illustrious
Society of Virtuosi which assembles at your houses and your goodness having
received M. Emmeriz and my self, very favorably at such times as we have
performd to entertain you either with discourse concerning it, or the sight of
some not inconsiderable effects of it (2).
iii
iv Foreword
REFERENCES
1. Lower R. The success of the experiment of transfusing the blood of one animal to
another. Philos Trans Royal Soc London 1666; 1:352.
2. Dennis J. A letter concerning a new way of curing sundry diseases by transfusion of
blood. Philos Trans Royal Soc London 1667; 2:489504.
3. Landsteiner J. Uber agglutinationserscheinungen normallen menschlichen blutes.
Wien Klin Wochenschr 1901; 14:11321134.
Preface
Preface
Human immunodeficiency virus (HIV) infection has become the most feared
complication of blood transfusion. Fear of acquired immunodeficiency syndrome
(AIDS) has triggered a series of radical changes in transfusion medicine. These
changes have focused mostly on the transmission of viruses. Other infectious and
noninfectious complications have been less well appreciated. One of the major
goals of this book is to examine transfusion risks in a broader context, and to
present some strategies to minimize these risks. A special effort was dedicated to
areas that have received less attention in recent years, particularly errors and
accidents, and the role of public health agencies as expressed by those in charge
of monitoring and regulation. Between 1990 and 1999, as risks of transmission
of HIV and hepatitis became relatively minuscule, there were 168 reports to FDA
of fatal hemolytic reactions due to ABO-incompatible transfusion through error.
This is an average of 17 cases a year, far exceeding deaths from transfusion-
associated infectious agents. The book also attempts to provide physicians and
other health care providers with the information necessary for appropriate coun-
seling of patients who may need, or who have received, blood transfusions and
for counseling of blood donors with abnormal screening test results.
The first chapter examines the contributions that blood donor screening
procedures make to transfusion safety. The remainder of the book is organized in
related sections to facilitate reference. Immunological complications discussed
include hemolytic transfusion reactions, other types of reactions, alloimmuniza-
tion, graft-versus-host disease, transfusion-related acute lung injury, and immuno-
modulation resulting from allogeneic transfusion. Also included is an overview
of transfusion-related errors and methods to prevent errors that may lead to
v
vi Preface
Jeanne V. Linden
Celso Bianco
Contents
Contents
I. Introduction
vii
viii Contents
A. General
B. Bacterial Infection
C. Viral Infections
D. Other
Index 607
Contributors
Contributors
Celso Bianco, M.D. Vice President, Medical Affairs, New York Blood Center,
New York, New York*
*Current affiliation: Executive Vice President, Americas Blood Centers, Washington, D.C.
xi
xii Contributors
Walter H. Dzik, M.D. Director, Blood Bank and Tissue Typing Labora-
tory, Beth Israel Deaconess Hospital, and Harvard Medical School, Boston,
Massachusetts
Jay S. Epstein, M.D. Director, Office of Blood Research and Review, Center
for Biologics Evaluation and Research, U.S. Food and Drug Administration,
Rockville, Maryland
Paul V. Holland, M.D. Medical Director and CEO, Sacramento Medical Foun-
dation Blood Centers, Sacramento, California
Jeanne V. Linden, M.D., M.P.H. Director, Blood and Tissue Resources, New
York State Department of Health, Wadsworth Center, Empire State Plaza, Albany,
New York
Maria Rios, Ph.D. Manager and Research Scientist, Scientific and Technolog-
ical Services Department, New York Blood Center, New York, New York
Iain J. Webb, M.D. Medical Director, Cell Manipulation and Gene Transfer
Laboratories, Dana-Farber Cancer Institute, and Instructor in Medicine, Harvard
Medical School, Boston, Massachusetts
Silvano Wendel, M.D. Medical Director, Blood Bank, Hospital Sirio Libanes,
So Paulo, Brazil
1
Impact of Blood Donor Screening
Procedures on Transfusion Safety
Steven Kleinman
University of British Columbia, Vancouver, British Columbia, Canada
I. INTRODUCTION
A major goal of transfusion medicine practice in the last decade has been to
reduce the risk of transfusion-transmitted infection to as low a level as possible.
In order to approach the desired level of zero risk from transfused allogeneic
blood, multiple layers of safety are needed. Methods utilized in attempting to
maximize safety from donated allogeneic units include donor-selection criteria,
donor-screening procedures, confidential unit exclusion (CUE) and telephone
callback procedures, laboratory testing, and modification of the blood unit after
collection, either by leukocyte removal or physicochemical procedures for patho-
gen inactivation.
This chapter will focus on donor selection and screening procedures, CUE
and callback procedures, product recall, and recipient notification in the event that
a potentially infectious unit has been transfused.
1
2 Kleinman
Timing Procedure
for the mentally retarded or prisons (10,11). In order to decrease the risk of human
immunodeficiency virus (HIV) transmission, blood collection agencies do not
collect from groups or institutions that are known to be comprised of male
homosexuals (12). Initially, these policies were instituted as primary protective
safety measures prior to the development of laboratory testing for the infectious
agents of hepatitis and acquired immunodeficiency syndrome (AIDS). However,
despite routine laboratory testing for these agents, these policies still remain
prudent in order to eliminate donors from settings where there is a high risk of
acute seronegative infection.
information to blood donors about the inappropriateness of HIV test seeking, and
some centers specifically ask donors whether they are donating to get an HIV test.
Nevertheless, it appears that more effective ways are needed to educate prospec-
tive donors that donating to obtain HIV antibody test results may endanger
transfusion recipients.
cially sensitive behavioral questions to the donor interview has continued to the
present day.
Health history interview questions designed to defer donors with increased
risk of infectious disease transmission can be broadly classified into several
categories. Table 2 indicates the diseases for which the various categories of
questions have been applied based upon known epidemiological risk factors.
Several methods have been utilized to conduct health history interviews:
these include the oral approach, the self-administered written approach, and
the combined approach. In the oral approach, a donor interviewer asks the
donor all health history questions and records the responses on the donor card.
In the self-administered approach, the donor checks off yes or no answers on the
donor card after reading each question. In the combined approach, the donor
may self-administer some questions and orally respond to others, or the
donor may self-administer all questions and then respond a second time to
selected questions orally presented by the donor interviewer. Regardless of the
approach utilized, if the donor gives answers that may potentially affect his or her
eligibility, the donor interviewer needs to elicit further information and to
document this on the donor history form.
Until recently, there was little standardization of donor questioning in
different blood centers in the United States. In 1991, a consortium of California
blood centers reported on their 3-year effort to design and implement a uniform
medical donor history card (57). The process used to generate the card was one
of consensus building among the medical directors of the various blood centers
with the final questions being approved by the Center for Biologic Evaluation and
Research (CBER) of the FDA. Limitations of this approach were an inability to
pretest the questions in donor populations prior to their implementation and the
reliance on medical directors rather than social scientists or communication
2. Review of Data
In addition to asking the right questions during the donor interview, it is also
necessary to obtain the right answers. Interviews with HIV- and HCV-seropositive
donors conducted after these donors have been notified of their test results show
that a high percentage of such donors will admit to a history of risky sexual
behavior or past intravenous drug use that they had denied prior to donating
(30,62). Recently, these data from seropositive donors have been complemented
by observations in a large geographically diverse donor population (9). In 1993,
REDS investigators mailed a 53-question optical scan format questionnaire to
50,162 allogeneic blood donors who successfully donated blood within the
previous 12 months at one of five participating REDS blood centers. Donors
were randomly sampled with the exception of oversampling of those younger
than 26 and those in minority ethnic and racial groups to compensate for their
lower proportional representation in the donor population. The questionnaire
contained items related to demographics, donation history, comprehension of
written donation literature, the use of CUE or callback procedures, sexual history,
injection drug use history, other history related to HIV risk, HIV test seeking,
donors knowledge about AIDS, and donors knowledge about donor eligibility
criteria. Multipart questions were used to determine the time intervals (corre-
sponding to intervals used in blood donor screening) in which events occurred:
these included ever, since 1977, within the past 12 months, and within the past
3 months. Questions pertaining to sexual behavior and injection drug use
Blood Donor Screening and Transfusion Safety 11
were preceded by a short statement explaining their purpose and the need for
truthful answers and giving the respondent permission to not answer any objec-
tionable questions.
Of 50,162 donors sampled, 34,726 (69.2%) responded and 98.1% of the
respondents answered all of the risk questions. The data were analyzed by using
sampling weights to adjust for differential sampling and/or response rates among
different demographic groups. A total of 1.9% of respondents reported at least one
behavioral risk that should have resulted in donor deferral. In 0.4%, this risk
occurred in the prior 3 months, a time frame compatible with acute seronegative
window period infection. These data demonstrate a low level of behavioral risk
that was not eliminated by donor questioning, laboratory testing, or CUE and
callback procedures. Given the strong psychological denial donors may have
about HIV risk behaviors or illicit activities, there is probably an inherent
limitation to the sensitivity that can be achieved by behavioral questioning (34).
Nevertheless, these data indicate that continued efforts to improve the sensitivity
of behavioral screening appear to be warranted (63).
3. Practical Aspects
Table 3 lists some of the important parameters that can be examined to potentially
increase the quality of the answers given in the donor health history interview.
Because the health history interview involves questions of a delicate nature,
it is important to ensure that the interview is conducted in a private and
confidential fashion. This can be difficult in the blood mobile setting. Two studies
have documented that the majority of donors felt that privacy during the history
interview was adequate (34,64). A study conducted by the American Red Cross
with 4651 donors at a variety of blood drives found that 74% of donors perceived
their privacy to be adequate at the health history station; this increased to 94%
when mobile visual partitions (standing screens) were used (64). These privacy
screens appeared to make a useful contribution to the donors perception of
privacy. Auditory privacy (the inability to hear another donors health history
interview) was considered as good to excellent by 92% of donors and increased
slightly to 97% when a device was used to mask extraneous noise. The second
temporary deferral which is removed 12 months after the last potential exposure.
These questions are:
In the past 12 months, have you had sex with a person who has HIV
infection or AIDS?
In the past 12 months, have you had sex with a person who currently or
previously used intravenous drugs?
(For Women) In the past 12 months, have you had sex with a man who has
had sex with another man (i.e., a man who is bisexual)?
In the past 12 months, have you had sex with a prostitute?
In the past 12 months, have you had sex with a person receiving clotting
factor concentrates?
In the past 12 months, have you had syphilis or gonorrhea?
In the past 12 months, have you had a blood transfusion?
In the past 12 months, have you had an accidental needlestick injury or a
blood splash to a mucous membrane or to nonintact skin?
At the conclusion of the medical history, donors must sign a consent that
specifically states that they understand that they should not donate blood if they
are at risk for HIV infection.
Two additional changes to HIV screening procedures have been imple-
mented since these comprehensive recommendations in 1992. In 1995, FDA
recommended that individuals who are inmates of correctional institutions and
individuals who have been incarcerated for more than 72 consecutive hours
during the previous 12 months be deferred for 12 months from their last date of
incarceration (70). FDA stated that this information could be provided to donors
through the predonation written material and did not indicate that this question
was required as part of the health history interview. In 1996, FDA recommended
that three questions be added to the direct questions on high-risk behavior to
exclude donors who are at increased risk for HIV-1 group O infection (71). One of
these questions relates to birth in or residence in Cameroon or surrounding West
African countries where HIV-1 group O infection has been identified. The others
refer to blood transfusion or medical treatment received in those countries and to
sexual contact with anyone who was born in or lived in those countries since
1977. These questions are similar in format to the type of geographic exclusion
questions for HIV-2 exposure that were made obsolete by the introduction of
HIV-2 antibody screening in 1992 (72).
the nurse to the donor such as Have you ever had sex with another male?) (6).
At the time there were no available data to indicate that more explicit questioning
might result in deferral of an increased number of high-risk persons; indeed, one
hypothesis was that direct questioning about a persons sexual orientation might
lead to untruthful answers because of concerns about maintaining the confidenti-
ality of information (6,12). A more direct approach also posed the possibility of
embarrassing or offending potential blood donors.
In the late 1980s, the donor interview process was reevaluated based upon
information obtained from interviews with HIV-seropositive donors and the
recognition of a much more open societal attitude towards discussing issues of
sexual orientation and sexual behavior. A retrospective study demonstrated that
asking donors direct oral questions about sexual behavior resulted in a fivefold
increase in HIV-related deferrals from 1.46 to 7.31 donors per month in one blood
center (73). A second blood center conducted a survey concerning donors
attitudes toward being asked such explicit sexual behavior questions. Of 1204
regular blood donors who responded, over 90% endorsed the use of these
questions, while only 1% felt that the questions might cause them to stop donating
(73). Subsequently, a prospectively designed study was performed in two U.S.
blood centers to address the effectiveness of donor-screening procedures, includ-
ing revised donor information brochures, the use of an AIDS information video,
and the use of a set of behavior-oriented direct questions asked orally by the
health historian. This study strongly suggested that the use of direct, behavior-
oriented oral questions led to a statistically significant increase in donors deferred
for HIV risk behavior (63). A second study, performed by the American Red
Cross, compared deferral rates in three groups, each of approximately 4000
donors, using direct behavior-oriented questions, indirect comprehension ques-
tions, and more generalized written interview questions; this study reached a
similar conclusion with regard to the benefit of direct oral questioning (74).
The current FDA and AABB recommendations suggest, but do not require, that
direct questions be asked orally (66,67).
With the implementation of anti-HIV testing in 1985, anti-HIV-positive
blood donors were identified, notified of their test results, and interviewed.
These interviews revealed that although many HIV-seropositive donors were
men who had sex with other men, they did not consider themselves to be at
risk for HIV infection (75). It became apparent that a person might respond
very differently if asked whether he were a member of a specific group (i.e.,
a homosexual man) as opposed to being asked if he had ever engaged in a
specific behavior (i.e., having sex with another male). The FDA therefore recom-
mended in September 1985 that any male who has had sex with another
male since 1977 should not donate blood (76). The recommendation emphasized
that individuals who had had only a single homosexual experience should refrain
from donating.
Blood Donor Screening and Transfusion Safety 17
Motivation (%)
CDC ARC
Reasons multicentera multicenter NIH
Denial of risk NA 61 26
Social pressure 27 29 15
Wanted to get test results 15 29 26
Previous HIV-seronegative test 9 NA NA
Positive test would stop blood from being transfused 10 NA 14
Misunderstood the pamphlet 7 NA 6
Other 32 NA 13
NA = Data not available in this format.
aSome donors gave more than one reason.
18 Kleinman
These data have demonstrated that seroconversion for HIV is highly likely to
occur within 6 months of HIV exposure (81) and that HIV nucleic acid cannot be
detected in HIV antibody-negative individuals from high-risk groups who are at
risk for latent HIV infection. Thus, HIV risk behaviors that can be defined as
ending at a specific point in time (i.e., sex with a particular person who
demonstrated HIV risk behaviors, an accidental exposure to blood, a blood
transfusion), should only defer a prospective donor until serological testing can
definitively prove that the individual is free of HIV infection. In order to allow
for outliers and to therefore be absolutely certain that the required time for
seroconversion has elapsed, a 12-month deferral interval for these behaviors has
been selected.
Other behaviors, such as past intravenous drug use or male-to-male sex
even once since 1977, still require a lifetime deferral. The unproven rationale for
a permanent deferral for a remote past history of male-to-male sex is that this
behavior represents a lifestyle choice, which may not be limited to a defined time
interval. To my knowledge, there are no data to support this contention and it
seems scientifically inappropriate to have differing deferral intervals for potential
homosexual and heterosexual exposure to HIV. With regard to intravenous drug
users, a permanent deferral seems appropriate give the higher seroprevalence of
most known infectious agents in this population and the concern that a past
injection drug user is more likely to be a carrier of other unidentified transfusion
transmissible agents.
As heterosexual transmission of HIV infection becomes more common in
the United States (82), it is to be anticipated that such a shift will be reflected in
the demographics of HIV-seropositive blood donors. Thus far, however, no
dramatic changes have occurred. In a CDC study of HIV-seropositive donors from
20 blood centers, the number of seropositive donors who acquired their HIV
infection by heterosexual exposure did not increase significantly when 1990
1991 data were compared to data from 19881989 (30). American Red Cross data
indicate that the rate of HIV-seropositive female donors has remained constant
from 1988 to 1992; however, a slightly decreasing rate of HIV-seropositive male
donors has resulted in an increased percentage of females among all HIV-
seropositive donors (83).
The risk of heterosexual spread of HIV has been addressed by several items
in the 1992 FDA recommendations (66). Several potential heterosexual risk
exposures are a cause for 12-month deferrals. These include sex with an intrave-
nous drug user, sex with a prostitute, sex with a bisexual man, and sex with a
person using clotting factor concentrates. In the early years of the AIDS epidemic,
donors who had immigrated to the United States from a country (e.g., Haiti and
sub-Saharan Africa countries) in which heterosexual activity played a major role
in transmission of HIV infection were permanently deferred (1,6). Questions
pertaining to geographic exclusion were continued until 1992 for sub-Saharan
Blood Donor Screening and Transfusion Safety 19
safety and can review HIV risk factor information with the donor. The physician
can obtain answers to explicit HIV risk questions from the donor. If the donor
admits to risk, he can be deferred. On the other hand, if the donor denies all HIV
risk, the physician will need to make a decision as to whether to believe the donor
or the third party. Unfortunately, there are no precise guidelines as to how such a
decision should be made. My belief is that when the third-party evidence is not
convincing and when the donor insists they have been truthful, the correct
procedure is to advise the donor that he is eligible to make future donations.
The donor will be reinterviewed and retested for HIV antibody on subsequent
donations, thereby providing some degree of confirmation of their safety.
One blood center reported their one-year experience with a protocol for
contacting the donor after third-party information was received (87). Eleven such
reports were received, 10 donors were interviewed, and 7 donors were determined
to be acceptable for future donation. The investigators stated that all contacted
donors were open, cooperative, and understanding of the blood centers position.
They concluded that the policy of confronting donors with third-party information
was effective in resolving these difficult donor suitability decisions.
B. Hepatitis
Federal guidelines for preventing transfusion-transmitted hepatitis were estab-
lished decades ago in the Code of Federal Regulations (CFR) (3). The current
regulations require the following deferral policies: (a) donors with a history of
viral hepatitis are permanently deferred; (b) donors with a history of close contact
with someone who has viral hepatitis are deferred for 12 months following their
last potential exposure; and (c) donors who have received a blood transfusion are
deferred for 12 months. [Although the CFR indicates a 6-month deferral for these
categories, subsequent FDA memos to blood establishments changed this deferral
Blood Donor Screening and Transfusion Safety 21
to 12 months (88). This change was based upon establishing consistency with
HIV deferral time intervals as well as the observed lag period to HCV seroconver-
sion following HCV exposure using first-generation HCV tests (89)]. In addition,
a person testing positive for HBsAg or known to have previously tested positive
for HBsAg is also permanently deferred as a blood donor.
Recently, FDA clarified that a history of viral hepatitis applies only to
clinical disease and that deferral is not required when the donors history is based
solely on a positive serological test result (i.e., anti-HBc or anti-HBs) that
indicates past exposure to HBV (90). Furthermore, the permanent deferral re-
quirement for a history of viral hepatitis no longer applies to an episode of viral
hepatitis occurring before the age of 10. (91). This recent change in FDA policy
is based upon epidemiological evidence that viral hepatitis in childhood, occur-
ring prior to the onset of sexual activity, is almost exclusively due to infection
with the hepatitis A virus (HAV) (92). Since HAV infection does not induce a
chronic carrier state, it is evident that persons who acquired HAV in the remote
past do not pose a hazard to transfusion recipients (93). Such reasoning raises the
issue of whether donors with a history of HAV at any age should be acceptable
as blood donors (94,95). Although medical knowledge supports the safety of
donations from these individuals, such a policy would be of limited value since
it would be difficult to prove definitively that a particular past episode of viral
hepatitis was due to HAV. Because of this problem in establishing accurate
diagnosis, it is still required that donors who present with a stated history of HAV
above age 10 be deferred.
In the past, blood collection agencies deferred all donors with a history of
jaundice (96). Currently the policy at most blood centers is to question the donor
concerning the etiology of the jaundice and to accept those donors whose history
of jaundice is related to the neonatal period or to obstructive biliary tract disease.
Other donor deferral criteria for potential hepatitis risk are based on the
known parenteral routes of spread of HBV and HCV. Persons who have ever
injected intravenous drugs by needle have long been deferred as blood donors due
to hepatitis risk; the importance of this criterion has been reemphasized with the
advent of the HIV epidemic.
Donors with a history of tattoos are deferred for 12 months because of the
possibility of hepatitis spread by contaminated needles (2). Donors who have had
needle exposure through ear piercing, skin piercing, acupuncture, or electrolysis
should also be considered for a possible 12-month deferral (2,6). In such cases it
should be evaluated whether these procedures were performed utilizing dispos-
able sterile needles as occurs in a professional setting; if so, deferral is not
necessary. If it cannot be verified that sterile technique was followed, then
consideration should be given to deferring such donors for 12 months.
Donors receiving hepatitis B immunoglobulin (HBIG) should be deferred
for 12 months following such administration because of the underlying hepatitis
22 Kleinman
B exposure risk. On the other hand, donors receiving hepatitis B vaccine do not
need to be deferred unless the vaccine was given for a recent exposure to HBV.
According to FDA regulations, donors who have had close contact with a
patient with acute viral hepatitis should be deferred for 12 months (88). Clearly,
sexual contact is included in this definition; it is less clear how to define
nonsexual close contact. One commonly used definition is the sharing of house-
hold, kitchen, or toilet facilities, as would occur with living in the same household
(6). In the case of HBV, this definition appears reasonable, in that it has been
demonstrated that HBV can rarely be transmitted from an acutely infected patient
to a household contact, probably through nonsexual contact with body fluids (97).
Data for nonsexual household transmission of HCV are more equivocal, and it is
unclear whether nonsexual contacts of such patients pose a risk to recipients (98).
Nevertheless, FDA requires that such prospective donors must be deferred.
On the other hand, persons who occasionally eat a meal or visit a patient who
has viral hepatitis may have little hepatitis risk; medical judgment should be
utilized to determine whether a particular donor with this type of history should
be deferred.
FDA requirements do not formally apply to sexual or close contacts of
asymptomatic or symptomatic chronic carriers of HBV and HCV. Given the high
rates of sexual and body fluid transmission of HBV, the same 12-month deferral
criterion should be applied to contacts of chronic HBV carriers. With regard to
HCV, data suggest that sexual transmission occurs with a low enough frequency
to make it safe to accept sexual partners of HCV carriers (99). Deferral policies
for close, nonsexual contacts of HCV carriers are more problematic given the
scarcity of reproducible clinical data; current data do not support the need to defer
such contacts (98).
C. Parasitic Diseases
Transfusion-transmitted malaria is common in some parts of the world but is rare
in the United States, occurring at an estimated rate of 0.25 cases per million
donated units for 19721988 (100,101). Policies for preventing such transmis-
sions rely on donor questioning during the health history interview.
The deferral criteria for malaria risk were revised by FDA in 1994 (102).
Travelers to a malaria endemic area are deferred for one year after their return to
the United States (provided they have not had malarial symptoms), whereas
immigrants from or residents of malarial endemic countries are deferred for 3
years after their departure from the endemic country. Donors with a history of
malaria are deferred for 3 years after becoming asymptomatic. Previous deferral
criteria in place from 1974 to 1994 had required that travelers to a malaria
endemic area be deferred for 3 years if they had received antimalarial prophylaxis
and for 6 months if they had not, because it was believed that prophylaxis might
Blood Donor Screening and Transfusion Safety 23
extend the latency period (3,101). The revised deferral criteria no longer use the
receipt of chemoprophylaxis as a determinant of the length of deferral.
Travelers to malarial endemic areas are deferred for one year because, if
infected, nonimmune persons will develop symptoms within this time frame.
The 3-year deferral period for immigrants from such areas is based on the premise
that such individuals may have partial tolerance to malarial parasites, thereby
resulting in the delay of malarial symptoms in an infected donor beyond one year
(3,100,101). The 3-year deferral for persons with a history of malaria is based on
the general consensus that Plasmodium falciparum organisms will be cleared
within 2 years and that P. vivax or P. ovale will be cleared within 3 years of the
resolution of symptoms in the vast majority of cases. This latter policy represents
a compromise between prevention of transmission and acceptable levels of donor
deferral, in that it is well known that a P. malariae chronic carrier state may persist
for decades, resulting in transfusion transmission many years after the resolution
of symptoms (103,104).
The CDC recently reported three cases of transfusion-transmitted malaria,
two of which were fatal (104a). In these two cases, the donors did not accurately
report their malaria risk information during the donor interview. These cases have
prompted an FDA review of malarial donor interview questions.
Chagas disease has been commonly transmitted by transfusion in Latin
America, but has only rarely been transmitted in this fashion in the United States
(105). Research studies conducted in the United States have attempted to identify
risk factors that might be associated with previous exposure to the causative
organism, Trypanosoma cruzi; these risk factors include birth in a country
endemic for Chagas disease, extended time spent in an endemic area, and lower
socioeconomic level (106). One institution with a large Hispanic donor popula-
tion has included Chagas risk questioning as part of its donor health history
interview and has performed investigational testing for T. cruzi antibody on
donors with affirmative answers prior to releasing blood for transfusion (5).
However, because of the low risk of documented transfusion-transmitted T. cruzi
infection in North America (four cases) and the low specificity of these proposed
questions, AABB-required health history questioning is confined to asking donors
whether they have ever had Chagas disease (2).
Babesiosis is a rare, malaria-like illness caused by a protozoan parasite,
Babesia microti, which invades human erythrocytes. Human transmission occurs
as a result of tick bites, most commonly in the summer months in particular
regions of the United States. Transfusion-transmitted babesiosis, although rare,
has been reported as a consequence of blood donation by asymptomatic carriers
(107,108). Strategies for eliminating transfusion risk by restricting donations
from endemic areas in the summer months or by questioning donors as to a
history of tick bites have been considered but have been judged to be largely
ineffective, although some centers nonetheless do avoid blood collection in highly
24 Kleinman
D. Creutzfeldt-Jacob Disease
Creutzfeldt-Jacob disease (CJD) is a rare, fatal, degenerative neurological disease
with a long asymptomatic latent period (111). The etiological agent is thought by
most experts to be a prion, although some favor a viral etiology (112). CJD has
been transmitted from human to human by the transplantation of dura mater, the
injection of pituitary-derived human growth hormone, and the reuse of EEG
electrodes; a single case of transmission by a corneal transplant has also been
reported (113). There are no reported cases of transmission by blood transfusion,
and epidemiological studies have not provided any evidence for such transmis-
sion (114,115). Nevertheless, because of the long incubation phase of the disease
(as demonstrated from growth hormone transmissions) and the inability of
conventional sterilization methods to inactivate the organism, the theoretical risk
of CJD transmission from asymptomatic donors to recipients of blood compo-
nents or plasma derivatives cannot be ruled out (116). Policies designed to prevent
this theoretical transfusion transmission of CJD have been adopted with regard to
donor deferral, product recall, and, in some cases, recipient notification (117120).
The initial concern for blood transfusion safety arose from the fact that
some persons who received therapeutic injections of cadaver-derived pituitary
human growth hormone developed CJD, thus raising the possibility that other
recipients of this hormonal product may have become asymptomatic chronic
carriers of the CJD agent (121,122). To eliminate possible risk to transfusion
recipients, FDA issued a 1987 recommendation that all prospective donors
be asked if they have ever received human growth hormone (117,118,122).
If the growth hormone was pituitary derived (i.e., the injection was given before
the availability of recombinant growth hormone in 1985), the donor was indefi-
nitely deferred.
In 1995 and 1996, FDA issued additional recommendations to further lower
the theoretical risk of transfusion-transmitted CJD by deferral of donors and recall
of previously acceptable donations (119,120). These recommendations included
the addition of questions about the receipt of a dura mater transplant and a family
history of CJD in a blood relative. Donors who respond affirmatively are
indefinitely deferred. A 9-month experience with CJD questioning at one large
blood center demonstrated that approximately one in 10,000 donors were deferred
for possible CJD risk, with 83% of the deferrals due to the donor having one blood
relative with CJD (123).
Since 90% of CJD cases are sporadic rather than familial, it should be
expected that a family history of one blood relative with CJD would be most
Blood Donor Screening and Transfusion Safety 25
likely due to the sporadic occurrence of the disease. Therefore, in 1996, FDA
policy for product recall for CJD was revised to indicate that product recall of
previously transfused single donor product and manufactured pooled plasma
derivatives was not necessary if a donor gave a history of only one family member
with CJD. Rather, the criteria for product recall would require two blood relatives
with CJD or genetic testing that was diagnostic of the familial variant (120).
In 1998, based on a review of international data, FDA amended its product recall
guidance to exclude recall of plasma-derivative products for donors with a history
of classical CJD or risk factors for classical CJD (120a).
F. Bacterial Disease
Another potential fatal complication of transfusion is bacterial infection (124).
Bacteria can be introduced into the donor unit at the time of collection if the donor
is bacteremic, if the skin site is not properly decontaminated, if there is an
undetected abscess adjacent to the phelbotomy site, or through introduction of a
small skin plug in the phlebotomy needle (124126). Examination of the skin at
the venipuncture site is conducted before phlebotomy, and strict requirements for
assuring the sterility of the site are adhered to by the phlebotomist (2,127). Blood
is not collected from persons who are febrile at the time of donation, who state
that they do not feel well, or who are taking systemic antibiotics (2). In the past,
blood centers have temporarily deferred donors with a history of dental proce-
dures for up to 72 hours because of the high frequency of postprocedure
bacteremia. These criteria have been liberalized with the recognition that many
26 Kleinman
other types of trauma to mucous membranes not evaluated at the time of donation
may also cause bacteremia (128). It has been recommended that donors need only
be deferred for 2472 hours after particular traumatic types of dental procedures,
such as root canals and tooth extractions; some blood centers no longer inquire
about recent dental procedures, and no such question is present on the AABB
uniform donor history card (58,124).
Unlike other infectious diseases, the risk of bacterial infection applies also
to candidates for autologous donation, as a result of the ability of gram-negative
rods to multiply at refrigerator temperatures and secrete endotoxin into the blood
bag (129). For this reason, autologous donors who are taking antibiotics or who
give a history of recent or concurrent medical procedures are evaluated for the
possibility of bacteremia and deferred accordingly.
are still permanently deferred. Donors with specific malignancies that have been
fully excised are not deferred (e.g., basal cell cancer of skin, cervical carcinoma
in situ), since the tumor is known to be low grade and not capable of hematoge-
nous spread.
names of donors deferred at any blood collection agency within the state (130).
The American Red Cross uses a national system in which donors who are entered
into specific categories of the deferral registry at an individual Red Cross region
also have their names included in a national registry (130,131). Because of donor
confidentiality concerns, donor deferral registries that extend beyond an individ-
ual blood center do not list the reason for donor entry into the registry.
Over the past 10 years, the size and complexity of donor deferral registries
have increased enormously. For example, the national component of the Ameri-
can Red Cross donor deferral registry contains over 20 separate deferral catego-
ries and over 300,000 donor names (130,131). It has been estimated that there
may be up to 3 million entries in donor deferral registries throughout the United
States (130). The problems associated with managing such expanded deferral
registries have also increased dramatically, creating a major source of difficulty
in adhering to FDA regulations. These regulations and blood center standard
operating procedures require that if a donor is listed in certain permanent deferral
categories on a donor deferral registry, additional donations from that individual
should not be distributed for transfusion. Difficulties in complying with this
requirement have resulted in numerous product recalls of subsequent units
donated by individuals who should have been placed on donor deferral registries
according to standard operating procedures. To my knowledge, there have been
no documented instances of these subsequently transfused units (which tested
infectious disease negative) causing adverse outcomes in recipients. These oper-
ational data suggest that donor deferral registries may not contribute significantly
to the enhancement of transfusion safety, given the other layers of safety that are
built into the system. A study to evaluate the usefulness of a regional donor
deferral registry demonstrated that 0.41% of donors who donated to a hospital-
based blood bank were later placed in that institutions donor deferral registry and
yet also donated to a regional blood center in the same geographic location (132).
These data can be interpreted to indicate that if donor deferral registries are
believed to be of value, their extension to regional registries may offer some
additional benefit.
Problems encountered in managing donor deferral registries include diffi-
culty in obtaining accurate information to uniquely identify a donor, the existence
of multiple records for the same donor due to conflicting information obtained on
separate donations, the need to move donors from one category of the registry to
another, and the fact that some information leads to permanent deferral while
other data only result in deferral if the phenomenon [e.g., a positive anti-hepatitis
B core (anti-HBc) test] occurs on two occasions (130).
Maintenance and continuous use of national or statewide donor deferral
registries is time-consuming, logistically complex, and expensive. Unfortunately
it has remained difficult to assess whether such massive efforts afford significant
increases in safety to the blood transfusion recipient.
Blood Donor Screening and Transfusion Safety 29
donors excluded their blood from transfusion, these two studies were able
to analyze whether donors who were demonstrated to seroconvert for HIV
(i.e., HIV-seronegative donation followed by HIV-seropositive donation) used
the CUE option on their preseroconversion sample. This is the most relevant
group for evaluation of CUEs effect on transfusion safety, since, unlike HIV-
seropositive units, these units would otherwise be transfused to recipients if the
CUE procedure were not in place (137,144). Investigators from the CDC com-
piled data on the use of CUE at the time of the preseroconversion donation of 322
HIV seroconverting donors at 40 blood centers from January 1987 through
December 1990 (144). They found that 3.4% of such donations were excluded;
however, 9 of the 11 exclusions came from one center (New York Blood Center),
which has consistently shown results for the CUE procedure that differ from the
remainder of the United States (54,135). If these data are eliminated, the data from
the remaining 39 centers indicate that 2 of 246, or 0.8%, of HIV-seroconverting
donors used this option. REDS analyzed the CUE process in 1.5 million donations
made at five blood centers in 1991 and 1992. These investigators found that 8%
of 169 HIV-seropositive donations were excluded and subsequently found that
6% (2 of 33) of HIV-seroconverting donors used this option on their pre-
seroconversion unit (145,146). Given the very low rate of HIV transmission by
transfusion and the low rate at which such seroconverting donors may choose the
CUE option, it has been estimated that this procedure may interdict one additional
HIV infectious unit per 7.14 million blood donations (146).
Despite its projected ineffectiveness in decreasing HIV risk from transfu-
sion, many still advocate retaining CUE as part of the donation process based on
other sources of data. Several studies have demonstrated that donors who use
CUE have higher rates of seropositivity for many infectious disease markers
(140,144,145,147) and that some individuals who use CUE admit to HIV risk
factors (139,142). The previously described anonymous mailed REDS survey
correlated the donors stated use of CUE (or telephone callback) with admitted
behavioral risk factors for infectious disease (9). This study found that all
behavioral risks, with the exception of injection drug use and history of transfu-
sion in the past year, were reported statistically significantly more frequently in
donors who used CUE. In aggregate, the relative prevalence of any reported risk
behavior in donors who used CUE was 7.6-fold greater than in those who did not
and was 9.4-fold greater for risk behaviors in the prior 3 months. However, CUE
was not used by the majority of donors who reported behavioral risk (low
sensitivity) and was used more frequently in donors without behavioral risk
(low specificity).
In 1992, FDA analyzed the then available data on CUE sensitivity and
specificity and stated that the CUE procedure was no longer mandatory, and its
use was left to the discretion of each individual blood center (66). Currently some
blood centers who previously used the CUE procedure have discontinued it.
Blood Donor Screening and Transfusion Safety 31
In summary, CUE has poor sensitivity and specificity but nevertheless may
prevent the transfusion of a very small number of units that cause infection
in recipients.
pertinent donor information that may assist with the recipient notification deci-
sion or message. In some cases, it may be helpful to the transfusion center
physician if donor center personnel are able to recall the donor in a timely manner
and obtain further clarifying history or pertinent serological testing.
Consider the case of a unit transfused from a donor who later reports HIV
risk factors. If such information is forwarded to a recipient, it would be expected
to cause anxiety. Furthermore, since HIV antibody testing will not be able to
definitively resolve the situation for several weeks to several months, this anxiety
may not be easily alleviated. In my opinion, the best way to handle such a
situation is for a blood center physician or designee to recontact the donor who
volunteered the information and attempt to get a specific and accurate history and,
if possible, a follow-up HIV test. This may allow the hospital transfusion service
medical director to better estimate the likelihood of risk to the recipient. Using
this information, the decision as to when and whether to notify the recipients
physician and/or the recipient and the details of the specific notification message
can be determined.
The American Association of Blood Banks has issued an Association
Bulletin to assist transfusion services in developing policies for physician and
recipient notification in cases in which the transfused component was obtained
from a donor who later revealed risk factors for or had a clinical diagnosis of
Creutzfeldt-Jacob disease (154). The situation with regard to these recipients is
problematic, since the possibility of transfusion transmission is only theoretical
and there is no diagnostic testing available for the recipient subsequent to
notification. The AABB states that the ultimate responsibility for deciding
to withhold information from a recipient rests with the recipients physician and
not with the medical director of the transfusion service or with the institution.
The AABB suggests that each hospital convene a committee, such as an Ethics
Committee or an Institutional Review Board, that could develop criteria for
informing or not informing patients of possible transfusion exposure to CJD.
These criteria would allow physicians at the institution to individualize decisions
for particular patients.
these specific blood components. These recipients, if still living and locatable,
can then be notified of their potential risk, usually by their physician. Laboratory
testing can be performed to determine whether infection occurred.
A generalized lookback program (also termed universal lookback) is a
program in which all recipients transfused within a designated time frame are
informed of their potential risk of infection from their transfusion. This program
can be implemented either through a public education campaign using the media
or communications to physicians or through direct mailings to recipients trans-
fused during the given time frame. This latter approach requires identification of
such recipients through hospital record searches.
The decision to perform targeted or generalized lookback programs for a
specific transmissible agent must consider multiple factors. Traditional public
health concerns, such as the yield and cost of the procedures, must be evaluated.
Public health authorities also need to consider lookback programs in the context
of more widespread screening programs to detect persons infected by routes other
than transfusion. In addition, an effective lookback program should ensure the
adequacy of diagnostic testing services, the adequacy of communication and
counseling resources for identified recipients, and medical follow-up for these
individuals. Another factor that may influence decisions concerning lookback
programs is the ethical premise that an individual has the right to know informa-
tion that might affect his or her future health. Additionally, liability concerns may
also influence institutional decision making.
In conjunction with considering these public health, ethical, and legal
issues, I believe that the answers to a critical set of medical questions are
important when formulating policies with regard to lookback for any transmissi-
ble agent. These questions are:
1. Is there evidence that the agent is transmissible by a particular blood
component?
2. Is there a diagnostic test to determine if infection has occurred in the
recipient?
3. Are there known modes of secondary transmission which can be
interrupted by the recipient having knowledge of his infection?
4. Are there medical means to monitor and assess the progression of the
disease in the recipient?
5. Is there treatment available?
These can be more concisely summarized into two broad considerations:
halting secondary spread of infection and the potential for intervening, or at least
monitoring, the natural history of the disease.
Testing of donated blood for anti-HIV, anti-HTLV, and anti-HCV has given
rise to the identification of blood donors who are HIV, HTLV, or HCV infected
and who have given previous, transfusable donations prior to the implementation
Blood Donor Screening and Transfusion Safety 35
of blood screening. These donors may have been infectious at the time of their
previous donation; therefore, recipients of these previous donations are at in-
creased risk of acquiring these infections. Currently, FDA requires targeted
lookback to be performed for donors identified as HIV antibody positive and has
established a rule requiring the transfusing institution to assume ultimate respon-
sibility for notification of the recipient or next of kin (66,156). A more limited
time frame of lookback, i.e., units transfused in the prior 3 months, is required
for donors identified as HIV p24 antigen positive (157). Although FDA does not
require targeted lookback for HTLV infection, the AABB has established such a
requirement (2,158). After several years of debate, FDA issued guidance requiring
that a targeted lookback program be conducted for recipients of blood products
from HCV antibodypositive donors. The specific requirements as to which
recipients must be informed are complex because of the evolution of HCV
screening and confirmatory tests (158). All transfusion recipients prior to 1992
are also part of a generalized lookback programs for HCV under the direction of
the U.S. Public Health Service (159).
VIII. CONCLUSION
Optimal donor-screening procedures represent a balance between maximizing
safety for both recipient and donor and minimizing the unnecessary deferral of
safe blood donors. Given the importance of recipient safety, decisions about
donor-screening policies tend to favor the use of less specific procedures in an
effort to enhance transfusion safety. In some cases, such enhancement can be
demonstrated, while in other cases it is inferred from indirect evidence and in still
other cases data may be completely lacking. Donor-screening policies can be
evaluated scientifically, providing that the inherent limitations of the methodol-
ogy used in this type of operational research are recognized. Additional attention
to quality assurance of donor-screening procedures is warranted due to the
difficult logistical situations that exist in some blood collection settings. Decisions
regarding product retrieval and recipient notification are complex and require
consideration of multiple factors.
REFERENCES
1. Kleinman S. Donor screening procedures and their role in enhancing transfusion
safety. In: Smith D, Dodd RY, eds. Transfusion-Transmitted Infections. Chicago:
American Society of Clinical Pathologists, 1991:207.
2. Klein HG, ed. Standards for Blood Banks and Transfusion Services. 17th ed.
Bethesda, MD: American Association of Blood Banks, 1996.
3. U.S. Department of Health and Human Services, Food and Drug Administration.
36 Kleinman
in United States plasma donors and in United States and French hemophiliacs.
Transfusion 1990; 30:780.
23. Dawson GJ, Lesniewski RR, Stewart JL, et al. Detection of antibodies to hepatitis
C virus in U.S. blood donors. J Clin Microbiol 1991; 29:551.
24. Mosley JW, Galambos JT. Viral hepatitis. In: Schiff L, ed. Disease of the Liver.
Philadelphia: JB Lippincott, 1975:500.
25. Kahn RA. Donor screening to prevent posttransfusion hepatitis. In: Keating LJ,
Silvergleid AJ, eds. Hepatitis. Washington, DC: American Association of Blood
Banks, 1981:99.
26. Taswell HA. Directed, paid and self donors. In: Clark GM, ed. Competition in Blood
Services. Arlington, VA: American Association of Blood Banks, 1987:137.
27. Strauss RG, Ludwig GA, Smith MV, et al. Concurrent comparison of the safety of
paid cytapheresis and volunteer whole-blood donors. Transfusion 1994; 34:116.
28. Huestis DW, Taswell HF. Donors and dollars (editorial). Transfusion 1994; 34:96.
29. Petersen LR, Doll LS. Human immunodeficiency virus type 1-infected blood
donors: epidemiologic, laboratory and donation characteristics. Transfusion 1991;
31:698.
30. Petersen LR, Doll LS, White CR, et al. Heterosexually acquired human im-
munodeficiency virus infection and the United States blood supply: considerations
for screening of potential blood donors. Transfusion 1993; 33:552.
31. Operskalski EA, Schiff ER, Kleinman SH, et al. Epidemiologic background of blood
donors with antibody to human T-cell lymphotropic virus. Transfusion 1989;
29:746.
32. Lee HH, Swanson P, Rosenblatt JD, et al. Relative prevalence and risk factors of
HTLV-I and HTLV-II infection in US blood donors. Lancet 1991; 337:1435.
33. Kolho EK, Krusius T. Risk factors for hepatitis C virus antibody positivity in blood
donors in a low-risk country. Vox Sang 1992; 63:192.
34. Doll LS, Petersen LR, White CR, et al. Human immunodeficiency virus type
1-infected blood donors: behavioral characteristics and reasons for donation. Trans-
fusion 1991; 31:704.
35. Leitman SF, Klein HG, Melpolder JJ, et al. Clinical implications of positive tests for
antibodies to human immunodeficiency virus type 1 in asymptomatic blood donors.
N Engl J Med 1989; 321:917.
36. Williams A, Kleinman S, Lamberson H, et al. Assessment of the demographic and
motivational characteristics of HIV seropositive blood donors. Paper presented at
the III International Conference on AIDS, Washington, DC, June 1987.
37. Mayo DJ. Evaluating donor recruitment strategies. Transfusion 1992; 32:797.
38. Williams AE, Watanabe K, Hansma D, et al. Prevalence and risk relationships of
blood donation incentives. Abstracts of FDA Workshop on incentives for volunteer
(nonrenumerated) donors. Bethesda, MD, September 1996.
39. Kleinman S. Donor incentives and motivation for donation: data from the CDC
sponsored multicenter study of HIV seropositive blood donors. Abstracts of FDA
Workshop on incentives for volunteer (nonrenumerated) donors. Bethesda, MD,
September 1996.
40. McFarland JG, Aster RH, Buggy BP. Paid time off for blood donationan incentive
to be less than truthful? Transfusion 1997; 37:447.
38 Kleinman
41. Kleinman SH, Shapiro A. The agreement to donate blood: Presenting information
to blood donors and obtaining consent. In: Smith DM, Carlson KB, eds. Current
Scientific/Ethical Dilemmas in Blood Banking. Arlington, VA: American Associa-
tion of Blood Banks, 1987:29.
42. Director of Recruitment Development, National Marrow Donor Program. Policy
regarding donor incentives at time of recruitment. Memorandum to donor center &
recruitment group coordinators and donor center medical directors, November 22,
1993.
43. Council of Europe. Guide to the Preparation Use and Quality Assurance of Blood
Components. Strasbourg: Council of Europe Press, 1992:12.
44. Mason JO. Alternative sites for screening blood for antibodies to AIDS virus. N Engl
J Med 1985; 313:1157.
45. Forstein M, Page PL, Coburn TJ. Alternative sites for screening blood for antibodies
to AIDS virus. N Engl J Med 1985; 313:1158.
46. Snyder AJ, Vergeront JM. Safeguarding the blood supply by providing opportunities
for anonymous HIV testing. N Engl J Med 1988; 319:374.
47. Lackritz EM, Kennedy MB, Doll LS, et al. Risk behaviors and test seeking among
HIV-1 positive blood donors. Transfusion 1995; 35(suppl):42S.
48. Williams AE, Thomson RA, Horton JA, Kleinman SH. Characterization of active
blood donors who recently donated blood primarily to receive an HIV test. Trans-
fusion 1995; 35(suppl):42S.
49. Korelitz JJ, Busch MP, Williams AE. Antigen testing for human immunodeficiency
virus (HIV) and the magnet effect: Will the benefit of a new HIV test be offset by
the numbers of higher-risk, test seeking donors attracted to blood centers? Transfu-
sion 1996; 36:203.
50. Stramer SL, Salemi BL, Sievert WS, et al. Evaluation of US blood donations testing
positive for HIV-1 p24 antigen. Transfusion 1996; 36(suppl):38S.
51. Dodd RY. Donor screening for HIV in the United States. In: Madhok R, Forbes CD,
Evatt BL, eds. Blood, Blood Products and AIDS. London: Chapman & Hall,
1987:143.
52. Director, Office of Biologics Research and Review, Food and Drug Administration.
Recommendations to decrease the risk of transmitting acquired immune deficiency
syndrome (AIDS) from blood donors. Memorandum to all establishments collecting
human blood for transfusion, March 24, 1983.
53. Seage III GR, Barry MA, Landers S, et al. Patterns of blood donations among
individuals at risk for AIDS, 1984. Am J Public Health 1988; 78:576.
54. Pindyk J, Waldman A, Zang E, et al. Measures to decrease the risk of acquired
immunodeficiency syndrome transmission by blood transfusion. Transfusion 1985;
25:3.
55. Perkins HA, Samson S, Busch MP. How well has self-exclusion worked? Transfu-
sion 1988; 28:601.
56. Busch MP, Young MJ, Samson SM, et al. Risk of human immunodeficiency virus
transmission by blood transfusions prior to the implementation of HIV antibody
screening in the San Francisco Bay Area. Transfusion 1991; 31:4.
57. Kolins J, Silvergleid AJ. Creating a uniform donor medical history questionnaire.
Transfusion 1991; 31:349.
Blood Donor Screening and Transfusion Safety 39
58. American Association of Blood Banks. Uniform donor history questionnaire. Asso-
ciation Bulletin #95-5, Bethesda, MD, June 2, 1995.
59. Sutor LJ, Sussman H, McCraw L, Fortenberry D. Regional standardization of the
blood donor medical history screening. Transfusion 1996; 36(suppl):88S.
60. Piliavan JA. Temporary deferral and donor return. Transfusion 1987; 27:199.
61. Sayers MH. Duties to donors. Transfusion 1992; 32:465.
62. Maclennan S, Barbara JA, Hewitt P, et al. Screening blood donations for HCV.
Lancet 1992; 339:131.
63. Mayo DJ, Rose AM, Matchett SE, et al. Screening potential blood donors at risk for
human immunodeficiency virus. Transfusion 1991; 31:466.
64. Kline L, Friedman LI, Dempesy D, et al. Assessment of blood donor privacy during
health history interviews. Transfusion 1996; 36:456.
65. Locke SE, Kowaloff HB, Hoff RG, et al. Computer-based interview for screening
blood donors for risk of HIV transmission. JAMA 1992; 268:1301.
65a. Kleinman S, Williams AE. Donor selection procedures: Is it possible to improve
them. Trans Med Rev 1998, 12:288.
66. Director, Center for Biologics Evaluation and Research. Recommendations for the
prevention of human immunodeficiency virus (HIV) transmission by blood and
blood products. Memorandum to all registered blood establishments, April 23, 1992.
67. American Association of Blood Banks. Statement and recommendations of the
American Association of Blood Banks regarding donor history questions. Memo-
randum to institutional members, July 29, 1989.
68. Mosley JW. Who should be our blood donors? Transfusion 1991; 31:684.
69. Westphal RG. Donors and the United States blood supply. Transfusion 1997;
37:237.
70. Director, Center for Biologics Evaluation and Research, Food and Drug Adminis-
tration. Recommendations for the deferral of current and recent inmates of correc-
tional institutions as donors of whole blood, blood components, source leukocytes,
and source plasma. Memorandum to all registered blood establishments, June 8,
1995.
71. Director, Center for Biologics Evaluation and Research, Food and Drug Adminis-
tration. Interim recommendations for deferral of donors at increased risk for HIV-1
group O infection. Memorandum to all registered blood and plasma establishments,
December 11, 1996.
72. OBrien RT, George JR, Holberg SD. Human immunodeficiency virus type 2
infection in the United States: epidemiology, diagnosis, and public health im-
plications. JAMA 1992; 267:2775.
73. Silvergleid AJ, Leparc GF, Schmidt PJ. Impact of explicit questions about high-risk
activities on donor attitudes and donor referral patterns-results in two community
blood centers. Transfusion 1989; 29:362.
74. Gimble JG, Friedman LI. Effects of oral donor questioning about high-risk behav-
iors for human immunodeficiency virus infection. Transfusion 1992; 32:446.
75. Schorr JB, Berkowitz A, Cumming PD, et al. Prevalence of HTLV-III antibody in
American blood donors. N Engl J Med 1985; 313:384.
76. Director, Office of Biologics Research and Review, Food and Drug Administration.
Revised definition of high risk groups with respect to acquired immunodeficiency
40 Kleinman
syndrome (AIDS) transmission from blood and plasma donors. Memorandum to all
registered blood establishments, September 3, 1985.
77. Sheppard HW, Dondero D, Arnon J, Winkelstein Jr W. An evaluation of the
polymerase chain reaction in HIV-1 seronegative men. J AIDS 1991; 4:819.
78. Eble BE, Busch MP, Khayam-Bashi H, et al. Resolution of infection status of
HIV-seroindeterminate and high-risk seronegative individuals using PCR and virus-
culture: absence of persistent silent HIV-1 infection in a high-prevalence area.
Transfusion 1992; 32:503.
79. Jackson JB. Human immunodeficiency virus (HIV) indeterminate Western blots and
latent HIV infection. Transfusion 1992; 32:497.
80. Centers for Disease Control and Prevention. Persistent lack of detectable HIV-1
antibody in a person with HIV infectionUtah, 1995. MMWR 1996; 45:181.
81. Busch MP, Satten GA. Time course of viremia and antibody seroconversion
following primary HIV infection: implications for management of exposed health
care workers. Am J Med 1997; 102(suppl 5B):117.
82. Brookmeyer R. Reconstruction and future trends of the AIDS epidemic in the United
States. Science 1991; 253:37.
83. U.S. Department of Health and Human Services, Public Health Service, Centers for
Disease Control and Prevention. National HIV Serosurveillance Summary. Results
Through 1992. Vol. 3, Atlanta, GA, 1993.
84. Schable C, Zekeng L, Pau CP, et al. Sensitivity of United States HIV antibody tests
for detection of HIV-1 group O infections. Lancet 1994; 334:1333.
85. Friedland GH, Saltzman BR, Rogers MF, et al. Lack of transmission of HTLV-
III/LAV infection to household contacts of patients with AIDS or AIDS related
complex with oral candidiasis. N Engl J Med 1986; 314:344.
86. American Association of Blood Banks. Management of hearsay information about
blood donors. Association Bulletin #93-4, Bethesda, MD, December 2, 1993.
87. Elliot D, Craig L, Price T. Resolving hearsay information received on a blood donor.
Transfusion 1995; 35(suppl):71S.
88. Director, Center for Biologics Evaluation and Research, Food and Drug Adminis-
tration. Revised recommendations for testing whole blood, blood components,
source plasma and source leukocytes for antibody to hepatitis C virus encoded
antigen (anti-HCV). Memorandum to all registered blood establishments, April 23,
1992.
89. Alter HJ, Purcell RH, Shih JW, et al. Detection of antibody to hepatitis C virus in
prospective followed transfusion recipients with acute and chronic non-A, non-B
hepatitis. N Engl J Med 1989; 321:1494.
90. Director, Center for Biologics Evaluation and Research, Food and Drug Adminis-
tration. Donor suitability related to laboratory testing for viral hepatitis and a history
of viral hepatitis. Memorandum to all registered blood establishments, December
22, 1993.
91. Director, Center for Biologics Evaluation and Research, Food and Drug Adminis-
tration. Exemptions to permit persons with a history of viral hepatitis before the age
of eleven years to serve as donors of whole blood and plasma: alternative proce-
dures, 21 CFR 640.120. Memorandum to all registered blood establishments, April
23, 1992.
Blood Donor Screening and Transfusion Safety 41
92. Trepo C. International forum: Should donors with a history of jaundice still be
rejected? Vox Sang 1981; 41:110.
93. Sheretz RJ, Russell BA, Reuman PD. Transmission of hepatitis A by transfusion of
blood products. Arch Intern Med 1984; 144:1579.
94. Alter HJ. Discussion: transfusion associated hepatitis. In: Polesky H, Walker RH,
eds. Safety in Transfusion Practices. Skokie, IL: College of American Pathologists,
1980:32.
95. Polesky HF, Hanson M. Tests for viral hepatitis markers in blood donors. In: Polesky
HF, Walker RH, eds. Safety in Transfusion Practices. Skokie, IL: College of
American Pathologists, 1980:17.
96. Aach RD. International forum: Should donors with a history of jaundice still be
rejected? Vox Sang 1981; 41:110.
97. Perillo RP, Gleb L, Campbell C, et al. Hepatitis B antigen, DNA polymerase activity,
and infection of household contacts with hepatitis B virus. Gastroenterology 1979;
76:1319.
98. Alter MJ. The detection, transmission, and outcome of hepatitis C virus infection.
Infect Agents Dis 1993; 2:155.
99. Seeff, LB, Alter HJ. Spousal transmission of the hepatitis C virus? Ann Intern Med
1994; 120:807.
100. Guerrero IC, Weniger BC, Schultz MG. Transfusion malaria in the United States,
19721981. Ann Intern Med 1983; 99:221.
101. Nahlen BL, Lobel HO, Cannon SE, Campbell CC. Reassessment of blood donor
selection criteria for United States travelers to malarious areas. Transfusion 1991;
31:798.
102. Director, Center for Biologics Evaluation and Research, Food and Drug Adminis-
tration. Recommendations for deferral of donors for malaria risk. Memorandum to
all registered blood establishments, July 26, 1994.
103. Sazama K. Prevention of transfusion-transmitted malaria: Is it time to revisit the
standards? Transfusion 1991; 31:786.
104. Bruce-Chwatt LJ. International Forum: Which are the appropriate modifications of
existing regulations designed to prevent transmission of malaria by blood transfu-
sion, in view of the increasing frequency of travel to endemic areas? Vox Sang 1987;
52:138.
104a. Centers for Disease Control and Prevention. Transfusion-transmitted malariaMis-
souri and Pennsylvania MMWR 1999; 48:253.
105. Wendal S, Gonzaga AL. Chagas disease and blood transfusion: a new world
problem? Vox Sang 1993; 64:1.
106. Leiby DA, Yund J, Read EJ, et al. Risk factors for Trypanosoma cruzi infection in
seropositive blood donors. Transfusion 1996; 36(suppl):57S.
107. Mintz ED, Anderson JF, Cable RG, et al. Transfusion-transmitted babesiosis: a case
report from a new endemic area. Transfusion 1991; 31:365.
108. Gerber MA, Shapiro ED, Krause PJ, et al. The risk of acquiring Lyme disease or
babesiosis from a blood transfusion. J Infect Dis 1994; 170:231.
109. Popovsky MA, Lindberg LE, Syrek AL, Page PL. Prevalence of babesia antibody
in a selected blood donor population. Transfusion 1988; 28:59.
110. American Association of Blood Banks Bulletin. Recommendations regarding babe-
42 Kleinman
121. Fradkin JE, Schonberger LB, Mills JL, et al. CJD in pituitary growth hormone
recipients in the United States. JAMA 1991; 265:880.
122. Holland PV. Why a new standard to prevent Creutzfeld-Jacob disease? Transfusion
1988; 28:293.
123. Kessler D, Bianco C. Donor deferrals related to Creutzfeldt-Jacob disease. Transfu-
sion 1996; 36(suppl):57S.
124. Goldman M, Blajchman MA. Blood product-associated bacterial sepsis. Trans Med
Rev 1991; 5:73.
125. Blajchman MA, Ali AM. Bacteria in the blood supply: an overlooked issue in
transfusion medicine. In: Nance SJ, ed. Blood Safety: Current Challenges. Bethesda,
MD: American Association of Blood Banks, 1992: 213.
126. Anderson KC, Lew MA, Gorgone BC, et al. Transfusion-related sepsis after
prolonged platelet storage. Am J Med 1986; 81:405.
127. American Association of Blood Banks. Bacterial contamination of blood products.
Association Bulletin #96-6, Bethesda, MD, August 7, 1996.
128. Ness PM, Perkins HA. Transient bacteremia after dental procedures and other minor
manipulations. Transfusion 1980; 20:82.
129. Richards C, Kolins J, Trindade CD. Autologous transfusion-transmitted Yersinia
enterocolitica. JAMA 1992; 268:1541.
130. Sherwood WC. Donor deferral registries. Transfusion Med Rev 1993; VII:121.
131. Grossman BJ, Springer KM. Blood donor deferral registries: highlights of a confer-
ence. Transfusion 1992; 32:868.
132. Grewal ID, Domen RE, Hirschler NV. The value of shared donor deferral registries.
Transfusion 1995; 35(suppl):66S.
133. Department of Health and Human Services, Food and Drug Administration. Addi-
tional recommendations for reducing further the number of units of blood and
plasma donated for transfusion or for further manufacture by persons at increased
risk of HTLV-III/LAV infection. Memorandum to all registered blood establish-
ments, October 30, 1986.
134. Ciavetta J, Nusbacher J, Wall A. Donor self-exclusion patterns and human im-
munodeficiency virus antibody test results over a twelve month period. Transfusion
1989; 29:81.
135. Loiacono BR, Carter GR, Carter CS, et al. Efficacy of various methods of confiden-
tial unit exclusion in identifying potentially infectious blood donations. Transfusion
1989; 29:823.
136. Busch MP, Perkins HA, Holland PV, et al. Questionable efficacy of confidential unit
exclusion (letter). Transfusion 1990; 30:668.
137. Petersen LR, Busch MP. Confidential unit exclusion: How should it be evaluated?
Transfusion 1991; 31:870.
138. Kean CA, Hsueh Y, Querin JJ, et al. A study of confidential unit exclusion.
Transfusion 1990; 30:707.
139. Wolles S, Galel S. Value of confidential unit exclusion. Transfusion 1993;
33(suppl):80S.
140. Kessler D, Valinsky JE, Bianco C. Sensitivity and Specificity of confidential unit
exclusion (CUE)-Does it work? Transfusion 1993; 33(suppl):35S.
141. Menitove JE, Lewandowski C, Ashworth LW, et al. Confidential unit exclusion
44 Kleinman
158. Center for Biologics Evaluation and Research. Food and Drug Administration.
Current Good Manufacturing Practice for Blood and Blood Components: (1) Quar-
antine and disposition of prior collections from donors with repeatedly reactive
screening tests for Hepatitis C virus (HCV); (2) Supplemental testing and the
notification of consignees and transfusion recipients of donor test results for anti-
body to HCV (anti-HCV). Draft Guidance for industry, Rockville, MD, June 1999.
159. U.S. Public Health Service. Briefing document on public health service options for
the identification of hepatitis C virus infection among prior transfusion recipients,
Washington, D.C., March 28, 1996.
2
Hemolytic Transfusion Reactions
I. HISTORICAL PERSPECTIVES
The concept of blood transfusion began with the description of the circulation of
blood by William Harvey in 1628. By the late 1660s, Jean Denis in France and
Richard Lowery in England were performing the first documented blood transfu-
sions into humans. Both used animals as the source of blood (1). In view of what
is now known about antispecies antibodies, it is not surprising that the first
description of a hemolytic transfusion reaction (HTR) was recorded by Denis in
1668 after giving a second infusion of calfs blood to a patient. Some of the same
symptoms described with this reactiondiaphoresis, back pain, tachycardia,
dyspnea, gastrointestinal distress, and dark urineremain classic findings for the
severe HTRs seen today (2). Deniss patient died after a third transfusion attempt,
and blood transfusions were subsequently banned by the French and British
medical societies (3).
In 1818, James Blundell rekindled interest in blood transfusion for use
primarily in the treatment of postpartum hemorrhage. He was the first to transfuse
blood from one human to another and to discourage the use of animals as donors
(1). The mortality rate of his patients was extremely high. However, because
Blundells patients were very ill, the role of incompatible blood and hemolytic
reactions in their clinical course is difficult to evaluate. Nevertheless, indiscrim-
inate use of blood transfusions by others during this period resulted in unaccept-
able rates of severe hemolytic reactions (4).
The modern era of blood transfusion began with Karl Landsteiners dis-
cover of the ABO blood group system in 1900. Initially, few people understood
47
48 Kicklighter and Klein
A. Immune-Mediated Versus
NonImmune-Mediated Hemolysis
An HTR occurs when a blood recipient has or subsequently develops antibodies
directed against antigens found on transfused red cells, resulting in accelerated
destruction of those cells. The recipient may mount an immune response if the
antigens on the infused cells are recognized as foreign. Antigenic stimulation may
50 Kicklighter and Klein
aseptic methods of collection and storage, and meticulous handling of blood, the
frequency of such reactions appears to be low.
Other mechanisms of nonimmune-mediated hemolysis include thermal,
mechanical, osmotic, and toxic damage to red cells from improper storage or
handling (2). Either overheating or freezing without an appropriate cryopreserva-
tive can result in significant hemolysis. Appropriate thermal monitoring should
be performed at every stage of blood collection, transport, storage, and infusion.
Blood should never be stored in an unmonitored refrigerator or warmed with a
device that has not been certified for this purpose. Transfusion of previously
frozen but inadequately deglycerolized red cells can result in osmotic hemolysis.
Osmotic hemolysis can also result from a simultaneous infusion of red cells and
a hypotonic solution. Mechanical trauma can lyse transfused red cells, especially
during infusion. For this reason, only pumps and other equipment that have been
properly validated for infusion of blood and blood components should be used.
Nonimmune-mediated hemolysis may occur in vivo posttransfusion,
from congenital defects of transfused red cells, such as glucose-6-phosphate
dehydrogenase (G6PD) deficiency, or from contamination with malarial parasites.
The latter is common in the developing world, but rare in donors screened in the
United States. Nonimmune-mediated hemolysis can occur with the infusion of
both allogeneic and autologous blood products. Since there are no screening tests
for nonimmune-mediated hemolysis that might occur in red cell units during
storage, visual inspection of each unit for hemolysis prior to issue and prior to
infusion is essential.
on the nature of the antibody and its ability to bind complement, the reaction can
result in intravascular or extravascular hemolysis.
Delayed hemolytic transfusion reactions (DHTR) are the result of immune-
mediated hemolysis that occurs days to weeks after the transfusion but is a direct
result of the transfusion. There is usually an anamnestic antibody response in
which antibody titers rise to detectable levels after transfusion of red cells
that bear an alloantigen to which the recipient has been previously sensitized.
Most DHTRs result in extravascular hemolysis. However, DHTRs can be associ-
ated with intravascular hemolysis, leading to life-threatening complications.
tering the blood should notify the patients primary physician immediately.
Treatment should be initiated as required clinically. The transfusion service
should be notified immediately and a transfusion reaction workup initiated.
Prompt blood bank evaluation can eliminate a potential second AHTR.
If there has been an identification error that involves mislabeled test samples or
misidentified blood components, a second patient could be in danger of receiving
a transfusion of incompatible blood. A patient who receives a random unit of
blood will have a one-in-three chance of a major ABO incompatibility (pa-
tients serum contains an ABO antibody to an antigen on the donors red cells)
that may result in a life-threatening transfusion reaction (10), and of these, about
a tenth are associated with a fatal outcome (24).
Fortunately, severe, acute life-threatening HTRs are rare. It has not been
possible to determine the most effective modes of therapy by means of controlled
studies. Therapy is supportive and must be directed towards correcting or
minimizing the pathophysiological events just described. Close monitoring and
aggressive support in an intensive care setting is indicated for all patients with
significant hemolysis.
Hypotension must be treated aggressively. Since the renal failure that may
accompany HTRs is thought to result from renal ischemia, therapy should be
directed toward the prevention of systemic hypotension and the maintenance of
renal cortical blood flow. If hypotension or shock can be adequately treated, renal
perfusion can usually be maintained and renal failure prevented (20).
One of the worst prognostic signs for the patient with an acute immune-
mediated HTR is the development of fulminant DIC. Fortunately, in most cases
the initiating immune hemolysis is short-lived, and few patients progress to DIC
(3). The optimal therapy for DIC precipitated by a transfusion reaction remains
controversial. Factors to consider when treating DIC include the severity of the
reaction, the antibody involved, the amount of incompatible blood infused, and
the patients clinical status. The infusion of fresh frozen plasma, cryoprecipitate,
and platelets may be necessary to stop hemorrhage and correct the coagulopathy.
The most severe HTRsthose that result in DICare associated with the
infusion of greater than 200 mL of ABO-incompatible blood. In the treatment of
severe AHTRs, Goldfinger has recommended the prophylactic use of heparin to
prevent DIC (22). However the efficacy of heparin in this setting is unknown.
Many of these patients may already have a source of active bleeding for which
the initial blood transfusion was ordered.
The response to all therapy should be closely monitored with frequent
laboratory and clinical assessment, and subsequent therapy should be guided by
both. The most useful measurements for ongoing hemolysis include hemoglobin
concentration, fractionated bilirubin, and lactic dehydrogenase (LDH); standard
measurements of coagulation and renal function are also indicated. The blood
bank can perform serological studies to determine whether an offending antibody
60 Kicklighter and Klein
reappear rapidly with subsequent stimulation, knowledge of prior red cell al-
loimmunization is essential to preventing the DHTR.
transfusion may be very difficult to distinguish from a sickle cell crisis. These
patients may become acutely ill as a result of a DHTR, with symptoms including
vaso-occlusive crisis, bone infarction, renal insufficiency, and profound anemia.
Initially, both DAT and IAT may be negative as a result of the clearance of
sensitized red cells. Elevated bilirubin lactate dehydrogenase (LDH), and urinary
hemoglobin, as well as other indications of hemolysis, may be present in both
conditions. If DHTR is not considered and the symptom complex is attributed to
a sickle cell crisis, these patients could continue to be transfused with antigen-
positive blood (31,32).
When fever without hemolysis occurs days to weeks after a transfusion,
transmission of an infectious agent or transfusion-associated graft-versus-host
disease should be considered (21). If delayed intravascular hemolysis of both
donor and recipient red cells is occurring, transfusion-induced malaria or babesi-
osis should be considered (33). The nonimmune-mediated hemolysis of trans-
fused G6PD-deficient red cells can also present with mild transient hemolysis
several days after their infusion, accompanied by mild jaundice and a two- to
threefold increase in bilirubin and LDH (3).
performed by the blood bank. Specimens sent to the blood bank should include
a coagulated and anticoagulated tube of blood, along with all suspected units, IV
tubing, and infusion sets. The patients blood samples must be drawn carefully to
avoid artifactual hemolysis. Symptoms that occur several hours after a transfusion
may be observed during the infusion of a subsequent unit in a patient receiving
multiple transfusions. The blood bank will perform a serological evaluation on all
suspected units.
The blood banks initial evaluation will include three steps: a clerical check,
an evaluation of hemolysis, and an evaluation for any evidence of serological
incompatibility. The clerical check will confirm the identity of the patients
sample and of the blood product(s), ensuring that there was no misidentification
that could have resulted in the patients receiving an incompatible unit.
Posttransfusion plasma will be examined for evidence of hemolysis. Intra-
vascular hemolysis of as little as 5 mL of blood usually raises the plasma
hemoglobin concentration to >50 mg/dL. This level of free plasma hemoglobin
should be rapidly and easily detectable by gross visual inspection of the
plasma/serum layer of a centrifuged blood specimen (6). Free hemoglobin is
usually cleared from the plasma in 512 hours. If the posttransfusion blood
sample is taken several hours after the transfusion, when the plasma could
possibly be cleared of hemoglobin, an assay for the presence of methemalbumin
might be useful (10). A posttransfusion urine sample can also be evaluated for
any evidence of hemoglobinuria. It is important that the urine is tested for free
hemoglobin and distinguished from hematuria and myoglobinuria.
Pre- and posttransfusion DATs are performed to check for serological
incompatibility (8). A positive posttransfusion DAT with a mixed-field appear-
ance is indicative of alloantibody coating antigen-positive transfused red cells.
Newly detected antibodies coating transfused cells in this setting are virtually
diagnostic of an immune basis for the hemolysis. Antibody may be demonstrable
in the patients serum using the IAT. However, the DAT may be positive days
before free antibody can be detected. It may be possible to identify the allo-
antibody by eluting it from the red cells.
When an AHTR is suspected after the three initial checks, additional
laboratory tests to determine the cause of the reaction may include confirmation
of ABO and Rh(D) types on pre- and posttransfusion samples as well as the
unit(s), repeat crossmatch, and other serological testing to detect alloantibodies
and/or incompatibility with donor units. If no clerical error has occurred, the
plasma is not grossly hemolyzed, and the DAT is negative or unchanged, an acute
HTR is extremely unlikely. However, if rapid hemolysis of all antibody-coated
red cells has occurred with rapid clearance of the free hemoglobin from the
plasma, or if antibody-coated red cells are removed rapidly from the circulation
by the reticuloendothelial system, immune-mediated AHTR is possible with a
negative DAT (21).
64 Kicklighter and Klein
If initial test results are unclear, additional confirmatory tests may be useful
to verify an AHTR. One should also determine whether a patient achieved the
expected hemoglobin rise after the transfusion: in a 70 kg adult, the hemoglobin
should increase by 1 g/dL per unit of red cells when measured 15 minutes after
the transfusion (34).
Serum bilirubin and LDH increase in both intravascular and extravascular
hemolysis. Rising unconjugated bilirubin may be detectable as early as one hour
postreaction, with peak levels occurring in 46 hours and disappearing in 24
hours if bilirubin excretion is normal. A serum haptoglobin level may be helpful
in suspected hemolysis, but a precipitous decrease in haptoglobin level tends to
occur very early and is not as reliable as hemoglobinemia. Visible hemoglobine-
mia develops after haptoglobin depletion, therefore little is gained by measuring
haptoglobin when hemolysis is already visible. The usefulness of a haptoglobin
level is also limited by the wide range of its normal values. Nonimmune-
mediated hemolysis will also lower haptoglobin concentrations.
When an immune-mediated DHTR has occurred, spherocytes observed on
the peripheral blood smear may be an early indication of red cell destruction (14).
A blood bank evaluation should be initiated in any case of suspected DHTR. If the
evaluation reveals that a positive DAT has developed, demonstrating the presence
of complement and/or antibody-coated red cells or the presence of previously
undetected red cell antibodies associated with a rapid disappearance of transfused
donor cells, then the diagnosis of a DHTR is made (21).
Characteristically, the DAT becomes positive a few days after transfusion
and remains positive until the incompatible red cells are eliminated. Typically,
antibody becomes detectable 47 days after transfusion and reaches a peak value
1015 days after transfusion (10). Antibody eluted from the red cells may help
identify the alloantibody when the antibody titer is low and difficult to detect in
the serum.
expression of an antigen. On the other hand, the major crossmatch confers the
ability to detect alloantibodies to uncommon antigens, which might not be present
on the standard panel of screening cells but are present on the donor red cells that
have been selected for the particular patient. The antibody screen and major cross
match complement one another. The FDA requires that 18 red cell antigens
representing the most clinically significant of the blood group systems be present
on reagent red cells used for antibody screening. In addition, most other high-
frequency antigens are present on reagent red cells (20). Only nine antibodies
66 Kicklighter and Klein
reactive to antigens not usually contained on commercial prepared red cell panel
cells have been the reported cause of HTRs. A critical review of these reports
reveals inadequate documentation in many instances (6). Under special condi-
tions, an electronic or computer crossmatch may substitute for serological
testing (36).
No in vitro test is capable of predicting the clinical significance of a red
cell antibody. However, several serological characteristics of red cell alloanti-
bodies correlate well with clinical consequences and form the basis for most of
our clinical judgment. The two serological characteristics that have proved most
helpful in predicting clinical significance are the antibody specificity, which is
determined by demonstrating reactivity with the corresponding red cell antigen,
and the antibodys in vitro reactivity at 3037C (37). Some antibodies are
invariably associated with hemolytic events, while others have never been
reported to cause untoward reactions in any patient. As previously discussed, the
severity of the hemolysis caused by these antibodies varies dramatically. Experi-
ence gained from transfusing patients in life-threatening situations when compat-
ible, antigen-negative blood supplies are unavailable or exhausted has illustrated
that anticipated hemolysis may not occur or may be minimal (38). This situation
is most likely to occur in the setting of a patient who has multiple red cell
alloantibodies. Although every effort should be made to transfuse these patients
with antigen-negative blood, in the emergency setting it may be necessary to
transfuse blood that is positive for the antigen(s) thought to be the least clinically
significant. In these situations, small aliquots should be infused slowly, with close
clinical supervision and careful monitoring for any evidence of hemolysis. In less
urgent situations where a negative crossmatch is difficult or impossible to obtain
and the red cell antibodies are of questionable clinical significance, in vivo
compatibility testing has been applied (15).
Just as the presence of an antibody does not necessarily mean that hemoly-
sis will occur, the absence of an antibody does not guarantee safety. Although
antibody screening and crossmatching with a recently obtained blood sample
identify most red cell alloantibodies, state-of-the-art serological techniques do not
identify all cases of incompatibility. Previously identified alloantibodies that may
be clinically significant should always be honored. Many clinically significant
antibodies become undetectable over time, only to reappear after transfusion of
red cells bearing the corresponding antigen. In recent studies by Ramsey et al.,
3035% of clinically significant red cell alloantibodies become undetectable on
follow-up screening within one year (with anti-Kidd undetectable in 59% of
follow-up screens and anti-C undetectable in 45% of follow-up screens), and
nearly 50% of all clinically significant alloantibodies became undetectable after
10 or more years (39).
Rarely, there will be cases where all evidence indicates immune-mediated
accelerated destruction of red cells in the absence of demonstrable antibodies
Hemolytic Transfusion Reactions 67
ACKNOWLEDGMENT
Much thanks to Ms. Jo Procter, M. Ed., MT, (ASCP) SBB, of the Transfusion
Medicine Department for her careful reading and thoughtful assistance with the
manuscript in draft.
REFERENCES
1. Wick MR. Blood transfusion. An historic and current perspective. Am J Clin Pathol
1989; 91(suppl):S2S6.
2. Beauregard P, Blajchman MA. Hemolytic and pseudo-hemolytic transfusion re-
actions: an overview of the hemolytic transfusion reactions and the clinical conditions
that mimic them. Transfusion Med Rev 1994; 8:184199.
3. Brecher ME, Taswell HF. Hemolytic transfusion reactions. In: Rossi EC, Simon TL,
Moss GS, eds. Principles of Transfusion Medicine. Baltimore: Williams and Wilkins,
1996:747763.
4. Oberman HA. The history of transfusion medicine. In: Petz LD, Swisher SN,
Kleinman S, et al., eds. Clinical Practice of Transfusion Medicine. New York:
Churchill Livingstone, Inc., 1996:1132.
5. Oberman HA. The crossmatch. A brief historical perspective. Transfusion 1981;
21:645651.
6. Shulman IA, Petz LD. Red cell compatibility testing: clinical significance and
laboratory methods. In: Petz LD, Swisher SN, Kleinman S, et al., eds. Clinical
Practice of Transfusion Medicine. New York: Churchill Livingstone, 1996:199244.
7. Daniels GL, Anstee DJ, Cartron JP, et al. Blood group terminology 1995. ISBT
working party on terminology for red cell surface antigens. Vox Sang 1995; 69:265
279.
8. Jeter EK, Spivey MA. Noninfectious complications of blood transfusion.
Hematol/Oncol Clin North Am 1995; 9:187204.
9. Pineda AA, Brzica SM Jr, Taswell HP. Hemolytic transfusion reaction. Recent
experience in a large blood bank. Mayo Clin Proc 1978; 53:387390.
10. Mollison PL, Engelfriet CP, Contreras M. Blood Transfusion in Clinical Medicine.
Oxford: Blackwell, 1993.
Hemolytic Transfusion Reactions 69
11. Lostumbro MM, Holland PV, Schmidt PJ. Isoimmunization after multiple trans-
fusions. N Engl J Med 1966; 275:141144.
12. Silberstein L, Spitalnik SL. Blood group antigens and antibodies. In: Rossi EC,
Simon TL, Moss GS, eds. Principles of Transfusion Medicine. Baltimore: Williams
and Wilkins, 1996:6787.
13. Pollack W, Ascari WQ, Crispen JF, et al. Studies on Rh prophylaxis. Rh immune
prophylaxis after transfusion with Rh-positive blood. Transfusion 1971; 11:340344.
14. Baron B, Baron J. Blood products and plasmapheresis. In: Hall JB, Schmidt GA,
Wood LDH, eds. Principles of Critical Care. New York: McGraw-Hill, 1992:
15. Silvergleid AJ, Wells RF, Hafleigh EB, et al. Compatibility test using 51-chromium-
labeled red blood cells in crossmatch positive patients. Transfusion 1978; 18:814.
16. Linden JV, Kaplan HS. Transfusion errors: causes and effects. Transfusion Med Rev
1994; 8:169183.
17. Walker RH. Special report: transfusion risks. Am J Clin Pathol 1987; 88:374378.
18. Linden JV, Paul B, Dressier KP. A report of 104 transfusion errors in New York state.
Transfusion 1992; 32:601606.
19. Sazama K. Reports of 355 transfusion-associated deaths: 1976 through 1985. Trans-
fusion 1990; 30:583590.
20. Schulman IA. Safety in transfusion practices. Red cell compatibility testing issues.
Clin Lab Med 1992; 12:685699.
21. Jenner PW, Holland PV. Diagnosis and management of transfusion reactions. In: Petz
LD, Swisher SN, Kleinman S, et al., eds. Clinical Practice of Transfusion Medicine.
New York: Churchill Livingstone, 1996:905927.
22. Goldfinger D. Acute hemolytic transfusion reactions: a fresh look at pathogenesis and
considerations regarding therapy. Transfusion 1977; 17:985987.
23. Davenport RD, Kunkel SL. Cytokine roles in hemolytic and nonhemolytic trans-
fusion reactions. Transfusion Med Rev 1994; 8:157168.
24. Murphy WG, McClelland DBL. Deceptively low morbidity from failure to practice
safe blood transfusion: an analysis of serious blood transfusion errors. Vox Sang
1989; 57:5962.
25. Boorman KE, Dodd BE, Loutit JF, et al. Some results of transfusion of blood to
recipients with cold agglutinins. Br Med J 1946; 1:751.
26. Fudenberg H, Allen FH. Transfusion reactions in the absence of demonstrable
incompatibility. N Engl J Med 1957; 256:11801184.
27. Moore BS, Taswell HF, Pineda AA, et al. Delayed hemolytic transfusion reactions.
Evidence of the need for an improved pretransfusion compatibility test. Am J Clin
Pathol 1980; 74:9497.
28. Pineda AA, Taswell HF, Brzica SM Jr. Delayed hemolytic transfusion reaction. An
immunologic hazard of blood transfusion. Transfusion 1978; 18:17.
29. Taswell HF, Pineda AA, Moore SB. Hemolytic transfusion reactions: frequency and
clinical and laboratory aspects. In: Bell CA, ed. A Seminar on Immune-Mediated Red
Cell Destruction. Washington, DC: American Association of Blood Banks, 1981:71
92.
30. Holland PV, Wallerstein RO. Delayed hemolytic transfusion reaction with acute renal
failure. J Am Med Assoc 1968; 204:10071008.
70 Kicklighter and Klein
31. Diamond WJ, Brown FL, Bitterman P, et al. Delayed hemolytic transfusion reaction
presenting as sickle cell crisis. Ann Intern Med 1980; 93:231233.
32. Milner PF, Squires JE, Larison PJ, et al. Posttransfusion crises in sickle cell anemia:
role of delayed hemolytic reactions to transfusion. South Med J 1985; 78:14621468.
33. Smith RP, Evans AT, Popovsky M, et al. Transfusion-acquired babesiosis and failure
of antibiotic treatment. J Am Med Assoc 1986; 256:27262727.
34. Wiesen AR, Hospenthal DR, Byrd JC, et al. Equilibrium of hemoglobin concentration
after transfusion in medical inpatients not actively bleeding. Ann Intern Med 1994;
121:278280.
35. Schulman IA. Controversies in red cell compatibility testing. In: Nance SJ, ed.
Immune Destruction of Red Blood Cells. Arlington, VA: American Association of
Blood Banks, 1989:171199.
36. Butch SH, Judd WJ, Steiner EA, et al. Electronic verification of donor-recipient
compatibility: the computer crossmatch. Transfusion 1994; 34:105109.
37. Garratty G. Factors affecting the pathogenicity of red cell auto- and alloantibodies.
In: Nance SJ, ed. Immune Destruction of Red Blood Cells. Arlington, VA: American
Association of Blood Banks, 1989, pp 109157.
38. Ramsey G, Cornell FW, Hahn L, et al. Red cell antibody problems in 1000 liver
transplants. Transfusion 1987; 27:552.
39. Ramsey G, Smietana SJ. Long-term follow-up testing of red cell alloantibodies.
Transfusion 1994; 34:122124.
40. Harrison CR, Hayes TC, Trow LL, Benedetto AR. Intravascular hemolytic trans-
fusion reaction without detectable antibodies: a case report and review of the
literature. Vox Sang 1986; 51:96101.
41. Jensen NJ, Crosson JT. An automated system for bedside verification of the match
between patient identification and blood unit identification. Transfusion 1996;
36:216221.
3
Other Reactions and
Alloimmunization
Christopher J. Gresens and Paul V. Holland
Sacramento Medical Foundation Blood Centers, Sacramento, California
I. INTRODUCTION
71
72 Gresens and Holland
observed that traditional platelet concentrates (PCs) were associated with a 9.3%
incidence of transfusion reactions (primarily rigors and/or fevers), whereas PCs
from which the buffy coats were removed prior to storage were associated with
an incidence of only 2.7% (p = 0.007) (13).
Fortunately, FNHTRs are almost never serious, although they can, in some
cases, be very uncomfortable to the patient. The fever usually responds to
antipyretics, such as acetaminophen. As with other acute transfusion reactions,
the transfusion should be discontinued and a work-up performed to rule out the
more serious types of reactions for which fever may be the harbinger (e.g., acute
HTRs, STRs, or TRALI). Most current literature states that the transfusion should
not be restarted with the same unit (14). In certain situations, however, such as
when the reaction is very mild, rapidly responds to antipyretics, and is definitively
shown to be unrelated to the aforementioned serious causes, some believe that
restarting the transfusion with the same unit may be acceptable (15).
The best means of preventing FNHTRs is to leukoreduce the cellular blood
component that is to be transfused. As mentioned previously, leukoreduction of
RBC units to levels of less than or equal to 5 108 leukocytes/unit eliminates
most FNHTRs (16), regardless of whether the reduction is done before or after
storage. Unfortunately, however, while poststorage leukoreduction (e.g., at the
bedside or in the lab immediately prior to issue) is reasonably effective at
preventing FNHTRs associated with RBC transfusions, it is less effective with
those due to platelet transfusions. This is because the aforementioned pyrogens
(IL-1, TNF-, etc.) reach much higher levels in nonprestorage-leukoreduced
platelet units than in similarly prepared RBC units, probably largely owing to the
increased storage temperature for platelets. Prestorage leukoreduction has an
obvious advantage here, in that it eliminates most of the leukocytes that would
otherwise produce pyrogens in vitro.
These days, plateletpheresis units are often collected in a manner that
automatically produces leukoreduced units and are therefore much less likely to
cause FNHTRs. Some blood centers have utilized this approach to set up a
virtually 100% leukoreduced plateletpheresis inventory, as the logistics needed to
accomplish this are not overly complicated. Prestorage leukoreduction of RBC
units and platelet concentrates, on the other hand, is a cumbersome process that
can be difficult to perform on every unit, and often necessitates the establishment
of a separate component inventory. For this reason, it has not usually been done
(until recently) on a very large scale. However, as the U.S. healthcare system
appears to be moving toward near-100% leukoreduced blood component inven-
tories, more and more blood collection facilities are gearing up their prestorage
leukoreduction processes for RBC and platelet concentrate units.
Generally, a patient will not receive leukoreduced cellular components
expressly for the prevention of FNHTRs unless he or she has had two such
documented reactions (assuming that the patient is not already receiving
74 Gresens and Holland
V. TRANSFUSION-ASSOCIATED CIRCULATORY
OVERLOAD
Rapid increases in blood volume are poorly tolerated in many patients with
cardiac, pulmonary, or renal failure, as well as in chronically anemic patients
with expanded plasma volumes and in very young or old patients (4,46). Mani-
festations of transfusion-associated circulatory overload (TACO) include dys-
pnea, rapid increases in systolic blood pressure, coughing, orthopnea, severe
headache, and peripheral edema. The reaction is not so much related to the blood
component itself as it is to the volume infused. In other words, the problem is
primarily a physiological one.
The incidence of TACO is difficult to determine, largely because of the
problem of underreporting. In a series from the Mayo Clinic, Popovsky and
Taswell (47) found that when the formal diagnosis of TACO was made by the
clinicians alone, only 1 in 3168 patients transfused with RBCs was reported as
being affected (as observed by a retrospective review looking at all transfusions
for a 7-year period). When they then set up a more structured consultation service,
that rate increased to 1 in 708. A recent study of elderly orthopedic surgery
patients revealed that over 1% of this population developed TACO. Not surpris-
ingly, the patients who developed TACO were older (mean of 84 vs. 77 years)
than those who did not, and each was in positive fluid balance (mean of 2480
mL) prior to administration of the offending transfusions (46). A final note
regarding the importance of TACO as a cause of patient morbidity and mortality
is that of 355 transfusion-associated deaths that occurred between 1976 and 1985,
39 were caused by acute pulmonary injury. This was the third most common cause
of death after acute hemolysis (158 deaths) and non-A, non-B hepatitis (42
deaths). Anaphylaxis accounted for 8 of the acute pulmonary injury-related
deaths, with the remaining 31 (9% of total deaths) caused by the acute onset of
pulmonary edema or respiratory insufficiency (30). While no distinction could be
made between TACO and TRALI as the cause of these, it is likely that at least
some (and perhaps many) were due to TACO.
Treatment of TACO is to stop the transfusion, sit the patient up as much as
possible, and give a diuretic (e.g., 40 mg intravenous furosemide for a typical
adult) and oxygen. In rare cases, such as when marked pulmonary edema occurs,
phlebotomy of 200400 mL whole blood, with the concomitant slow transfusion
of packed RBCs, may be useful (this is actually faster than plasmapheresis).
Prevention may be accomplished by transfusing the at-risk patient with very
small volumes of the required component, as slowly as possible. Generally, the
Other Reactions and Alloimmunization 79
transfusion rate should not exceed 1 mL per kilogram body weight per hour
(18,48). Also, consideration should be given to administering a diuretic prior to
the transfusion of an at-risk individual. In rare cases, partial RBC exchange
(i.e., removing whole blood and replacing with packed RBCs) may be utilized in
order to reduce the patients plasma volume while increasing his or her hemo-
globin concentration.
REFERENCES
1. Heddle NM, Klama LN, Griffith L, Roberts R, Shukla G, Kelton JG. A prospective
study to identify the risk factors associated with acute reactions to platelet and red
cell transfusions. Transfusion 1993; 33:794797.
2. Heddle NM, Klama L, Singer J, Richards C, Fedak P, Walker I, Kelton JG. The role
of plasma from platelet concentrates in transfusion reactions. N Engl J Med 1994;
331:625628.
3. Payne R. The association of febrile transfusion reactions with leuko-agglutinins. Vox
Sang 1957; 2:233241.
4. Litty C. A review: Transfusion reactions. Immunohematology 1996; 12:7279.
5. Meryman HT, Hornblower M. The preparation of red cells depleted of leukocytes.
Transfusion 1986; 26:101106.
6. Mangano MM, Chambers LA, Kruskall MS. Limited efficacy of leukopoor platelets
for prevention of febrile transfusion reactions. Am J Clin Pathol 1991; 95:733738.
7. Dzik WH. Is the febrile response to transfusion due to donor or recipient cytokines?
Transfusion 1992; 32:594.
8. Muylle L, Joos M, Wouters E, De Bock R, Peetermans ME. Increased tumor necrosis
factor alpha (TNF-alpha), interleukin 1 (IL-1), and interleukin 6 (IL-6) levels in the
plasma of stored platelet concentrates: relationship between TNF-alpha and IL-6
levels and febrile transfusion reactions. Transfusion 1993; 33:195199.
9. Ferrara JLM. The febrile platelet transfusion reaction: a cytokine shower. Transfusion
1995; 35:8990.
10. Aye MT, Palmer DS, Giulivi A, Hashemi S. Effect of filtration of platelet concentrates
on the accumulation of cytokines and platelet release factors during storage. Trans-
fusion 1995; 35:117124.
11. Stack G, Snyder EL. Cytokine generation in stored platelet concentrates. Transfusion
1994; 34:2025.
12. Federowicz I, Barrett BB, Anderson JW, Urashima M, Popovsky MA, Anderson KC.
A characterization of reactions after transfusion of cellular blood components that are
white cell reduced before storage. Transfusion 1996; 36:2128.
84 Gresens and Holland
13. Muylle L, Wouters E, Peetermans ME. Febrile reactions to platelet transfusion: the
effect of increased interleukin 6 levels in concentrates prepared by the platelet-rich
plasma method. Transfusion 1996; 36:886890.
14. Widmann FK. Controversies in transfusion medicine: Should a febrile transfusion
reaction occasion the return of the blood component to the blood bank? Pro.
Transfusion 1994; 34:356358.
15. Oberman HA. Controversies in transfusion medicine: Should a febrile transfusion
response occasion the return of the blood component to the blood bank? Con.
Transfusion 1994; 34:353355.
16. Ryden SW, Oberman HA. Compatibility of common intravenous solutions with CPD
blood. Transfusion 1975; 15:250255.
17. Kevy S, Schmidt PJ, McGinniss MH, Workman WG. Febrile nonhemolytic trans-
fusion reactions and the limited role of leukoagglutinins in their etiology. Transfusion
1962; 2:716.
18. Vengelen-Tyler V, ed. Noninfectious complications of blood transfusion. In: Techni-
cal Manual. 12th ed. Bethesda, MD: American Association of Blood Banks,
1996:543562.
19. Vesilind GW, Simpson MB, Shifman MA, Colman RE, Kao KJ. Evaluation of a
centrifugal blood cell processor for washing platelet concentrates. Transfusion 1988;
28:4650.
20. Silvergleid AJ, Hafleigh EB, Harabin MA, Wolf RM, Grumet FC. Clinical value of
washed-platelet concentrates in patients with non-hemolytic transfusion reactions.
Transfusion 1977; 17:3337.
21. Sandler SG, Eckrich R, Malamut D, Mallory D. Hemagglutination assays for the
diagnosis and prevention of IgA anaphylactic transfusion reactions. Blood 1994;
84:20312035.
22. Poothullil J, Shimizu A, Day RP, Dolovich J. Anaphylaxis from the product(s) of
ethylene oxide gas. Ann Intern Med 1975; 82:5860.
23. Branch DR, Gifford G. Allergic reaction to transfused cephalothin antibody. JAMA
1979; 241:495496.
24. Stern A, van Hage-Hamsten M, Sondell K, Johansson SGO. Is allergy screening of
blood donors necessary? Vox Sang 1995; 69:114119.
25. Michel J, Sharon R. Non-hemolytic adverse reaction after transfusion of a blood unit
containing penicillin. Br Med J 1980; 1:152153.
26. Take H, Tamura J, Sawamura M, Murakami H, Naruse T, Tsuchiya, Miyawaki S,
Hirabayashi H. Severe anaphylactic transfusion reaction associated with HLA-
incompatible platelets. Br J Haematol 1993; 83:673674.
27. Sano H, Koga Y, Hamasaki K, Furuyama H, Itami N. Anaphylaxis associated with
white-cell reduction filter. Lancet 1996; 347:1053.
28. Bjerrum OJ, Jersild C. Class-specific anti-IgA associated with severe anaphylactic
transfusion reactions in a patient with pernicious anemia. Vox Sang 1971; 21:411424.
29. Pineda A, Taswell HF. Transfusion reactions associated with anti-IgA antibodies:
Report of four cases and review of the literature. Transfusion 1975; 15:1015.
30. Sazama K. Reports of 355 transfusion-associated deaths: 1976 through 1985. Trans-
fusion 1990; 30:583590.
31. Lippman M, Rumley W. Medical emergencies. In: Dunagan WC, Ridner ML, eds.
Other Reactions and Alloimmunization 85
Manual of Medical Therapeutic. 26th ed. Boston: Little, Brown and Company,
1989:482503.
32. Shirey RS, Ness PM. Alloimmunization to blood group antigens. In: Anderson KC,
Ness PM, eds. Scientific Basis of Transfusion Medicine: Implications for Clinical
Practice. Philadelphia: W.B. Saunders Company, 1994:507516.
33. Rosse WF, Gallagher D, Kinney TR, Castro O, Dosik H, Moohr J, Wang W, Levy
PS, and the Cooperative Study of Sickle Cell Disease. Transfusion and alloimmuniza-
tion in sickle cell disease. Blood 1990; 76:14311437.
34. Hoeltge GA, Domen RE, Rybicki LA, Schaffer PA. Multiple red cell transfusion and
alloimmunizationexperience with 6996 antibodies detected in a total of 159,262
patients from 1985 to 1993. Arch Pathol Lab Med 1995; 119:4245.
35. Lostumbo MM, Holland PV, Schmidt PJ. Isoimmunization after multiple trans-
fusions. N Engl J Med 1966; 275:141144.
36. Coles SM, Klein HG, Holland PV. Alloimmunization in two multitransfused patient
populations. Transfusion 1981; 21:462466.
37. Wayne AS, Kevy SV, Nathan DG. Transfusion management of sickle cell disease.
Blood 1993; 81:11091123.
38. Diamond WJ, Brown FL, Bitterman P, Klein HG, Davey RJ, Winslow RM. Delayed
hemolytic transfusion reaction presenting as sickle-cell crisis. Ann Intern Med 1980;
93:231234.
39. Milner PF, Squires JE, Larison PJ, Charles WT, Krauss JS. Posttransfusion crisis in
sickle cell anemia: role of delayed hemolytic reactions to transfusion. Southern Med
J 1985; 78:14621469.
40. The Trial to Reduce Alloimmunization to Platelets Study Group. Leukocyte reduction
and ultraviolet B irradiation of platelets to prevent alloimmunization and refractori-
ness to platelet transfusions. N Engl J Med 1997; 337:18611869.
41. Williamson LM, Wimperis JZ, Williamson P, Copplestone JA, Gooi HC, Morgenstern
GR, Norfolk DR. Bedside filtration of blood products in the prevention of HLA
alloimmunizationA prospective randomized study. Blood 1994; 83:30283035.
42. von dem Borne AEGK, Simsek S, van der Schoot CE, Goldschmeding R. Platelet
and neutrophil alloantigens: their nature and role in immune-mediated cytopenias. In:
Garratty G, ed. Immunobiology of Transfusion Medicine. New York: Marcel Dekker
Inc., 1994:149171.
43. Puig N, de Haas M, Kleijer M, Montoro JA, Perez A, Villalba JV, Gomez I, von dem
Borne AEJK. Isoimmune neonatal neutropenia caused by FcgammaRIIIb antibodies
in a Spanish child. Transfusion 1995; 35:683687.
44. Van Buren NL, Stroncek DF, Clay ME, McCullough J, Dalmasso AP. Transfusion-
related acute lung injury caused by an NB2 granulocyte-specific antibody in a patient
with thrombotic thrombocytopenic purpura. Transfusion 1990; 30:4245.
45. Popovsky MA, Chaplin HC, Moore SB. Transfusion-related acute lung injury: a
neglected, serious complication of hemotherapy. Transfusion 1992; 32:589591.
46. Popovsky MA, Audet AM, Andrzejewski Jr. C. Transfusion-associated circulatory
overload in orthopedic surgery patients: a multi-institutional study. Immunohemato-
logy 1996; 12:8789.
47. Popovsky MA, Taswell HF. Circulatory overload: an underdiagnosed consequence of
transfusion. Transfusion 1985; 25:469.
86 Gresens and Holland
48. Marriott HL, Kekwick A. Volume and rate in blood transfusion for the relief of
anemia. Br Med J 1040; 1:10431046.
49. Kim HC, Dugan NP, Silber JH, Martin MB, Schwartz E, Ohene-Frempong K, Cohen
AR. Erythrocytapheresis therapy to reduce iron overload in chronically transfused
patients with sickle cell disease. Blood 1994; 83:11361142.
50. Propper RD, Button LN, Nathan DG. New approaches to the transfusion management
of thalassemia. Blood 1980; 55:5560.
51. Hall TL, Barnes A, Miller JR, Bethencourt DN, Nestor L. Neonatal mortality
following transfusion of red cells with high plasma potassium levels. Transfusion
1993; 33:606609.
52. Brown KA, Bissonnette B, MacDonald M, Poon AO. Hyperkalemia during massive
blood transfusion in paediatric craniofacial surgery. Can J Anaesth 1990; 37:401408.
53. Kohan DE. Fluid and electrolyte management. In: Dunagan WC, Ridner ML, eds.
Manual of Medical Therapeutics. 26th ed. Boston: Little, Brown and Company,
1989:5271.
54. Linden JV, Kaplan HS, Murphy MT. Fatal air embolism due to postoperative blood
recovery. Anesthesia Analgesia 1997, 84:422426.
55. Hume HA, Popovsky MA, Benson K, Glassman AB, Hines D, Oberman HA,
Pisciotto PT, Anderson KC. Hypotensive reactions: a previously uncharacterized
complication of platelet transfusion? Transfusion 1996; 36:904909.
56. Owen HG, Brecher ME. Atypical reactions associated with use of angiotensin-
converting enzyme inhibitors and apheresis. Transfusion 1994; 34:891894.
57. Moore AB. Hypotensive reactions: Are they a new phenomenon? Are they related
solely to transfusion of platelets? Does filtration of components play a role? Trans-
fusion 1996; 36:852853.
58. Adverse ocular reactions following transfusionUnited States, 19971998. MMWR
1998; 47:4950.
4
Managing Error for
System Improvement
Harold S. Kaplan
New York-Presbyterian Hospital, New York, New York
James B. Battles
University of Texas, Southwestern Medical Center, Dallas, Texas
The safety of blood transfusion has long been an area of concern and study (1).
However, with the intense public and legislative scrutiny following the discovery
of the potential for human immunodeficiency virus (HIV) transmission by blood
transfusion, there has been an exceptional increase in the investigative and
regulatory interest directed at a broad range of blood safety issues.
In an effort to minimize undesirable variations in the manufacturing
aspect of transfusion medicine, i.e., the recruitment, collection, testing, and
processing of units of donated blood, there is an increased regulatory emphasis
on rigorous process control coupled with training to protocols of standard
operating procedures (2). This approach, contained within the Current Good
Manufacturing Practices (CGMPs), clearly contributes to more reliable processes.
However, even with the long-established CGMPs in the biopharmaceutical man-
ufacturing industry, there is a documented persistence of human error even with
strict adherence to CGMPs (3). Similar problems are seen with the process of
transfusion to the intended recipient.
87
88 Kaplan and Battles
A. Observation/Audit
One approach has been direct observation by skilled observers in the actual
operating environment. This is a well-established way to address human error in
its actual setting (4). Shulman and coworkers (5) developed a multidisciplinary
team approach to quality assurance and improvement directed at reducing pa-
tient identification errors by improving compliance with standard procedures.
The program involved periodic concurrent audits, which included direct observa-
tion of procedures. With feedback of deviations from protocol and active educa-
tional efforts, adherence to patient identification protocols improved gradually
from 50% during a pilot study to nearly 100% by the 125th audit (5). Although
the authors found in-service education to be effective in increasing compliance
with protocol and reducing the risk of error, observation may itself alter the
circumstances studied, as may observer error and limitation in controlling all
relevant variables. In addition, the enduring effect of such improvement may be
difficult to maintain given employee turnover and the need for sustained inten-
sive effort.
B. Accident Analysis
The second approach to error identification and prevention is the analysis of
accident data. This approach has been an important source of information used
by the National Transportation Safety Board (NTSB) in aviation (4). However,
hindsight bias and incomplete data lead to distortion. Additionally, although
human error has often been identified as a cause of an accident, little insight has
been provided as to why an error occurred.
Despite these limitations, analysis of accident data has been an important
source of information in blood transfusion. In 1975, the Food and Drug Admin-
istration (FDA) first mandated the reporting of transfusion-associated fatalities,
and by 1980, the Health Care Finance Administration agreed to investigate these
fatalities in greater depth. The file of these reports is accessible under the Freedom
of Information Act. This database has been extensively studied by a number of
investigators (610).
Mummert and Tourault (11) reviewed 150 transfusion-associated fatalities
reported to the FDA from 1990 to 1992. They concluded that nearly one third of
these fatalities could have been prevented by adherence to proper procedure.
Interestingly, a failure to follow procedures is also responsible for one third of
Managing Error for System Improvement 89
major air carrier accidents. However, as pointed out by Nagle (4), even with
categorization of error data, if it is not known why someone failed to follow
standard procedures, development of an effective preventive strategy remains
problematic. In this regard, Nagle has stressed the need for a model of human
error to be used in conjunction with error data collection and classification.
Mummert and Tourault also noted that improper transfusion of ABO-
incompatible red blood cells due to error continues to be a primary cause of
preventable death. They also reported that failure to identify a reaction in progress
contributed to many of the fatalities. Again, in analogy with other error-critical
areas, such as nuclear power and aviation, delay in detecting a problem, im-
properly identifying its cause, and delay in implementing corrective actions are
recognized as critical issues (12).
The management of error to limit adverse outcomes or unplanned effects is
now recognized to be of fundamental importance in system design and training
in error-critical activities, particularly since the Three Mile Island accident (12).
Although the importance of this has also been appreciated in transfusion medi-
cine, its proper emphasis has not been carried through to all critical operational
areas. In some cases analyzed by Mummert and Tourault (11), signs or symptoms
were treated, but the transfusion was not identified as the cause and was
continued. These authors also reported that in several cases signs such as
hemoglobinuria were noted without being recognized as a transfusion reaction.
In an extensive analysis of fatalities reported to the FDA over a 10-year
period, Sazama (10) reviewed 355 reports and studied 256 (99 did not involve
transfusion, 68 related to transfusion-associated hepatitis, 3 related to transfusion-
associated acquired immune deficiency syndrome, and one report recorded could
not be located). Acute hemolytic transfusion reaction resulting from ABO incom-
patibility accounted for the majority of fatalities. Sazama found one third of all
deaths and two thirds of incompatible red cell transfusions to be attributable to
error and therefore preventable. She estimated that 124 fatal ABO errors occurred
in approximately 100,000,000 transfusions, or 1/800,000 units. Sazama also
reported that 5 of the 26 hepatitis B deaths resulted from error, with three
seropositive units labeled properly but released in error, and two seropositive
units labeled improperly as negative. Additionally, of 12 reported donor deaths,
one related to an O-positive plasma donor who received A red cells from another
donor. Sazama determined that errors leading to fatality were most often mana-
gerial or system errors, rather than isolated human error.
In a 2-year period from 1990 through 1991, transfusion-related incidents
reported to the New York State Department of Health (NYSDOH) also included
errors that as precursor events could have but did not necessarily result in harm
(13). The reported incidence of administration of the wrong unit of blood, or of
blood given to the wrong recipient, was estimated at 1/12,000 units transfused.
Of these mistakes, ABO-incompatible red cell transfusions had an estimated
90 Kaplan and Battles
C. Simulation
A third method of error analysis for development of prevention strategies is the
study of error in the laboratory and by simulation. This approach has also proven
useful since laboratory circumstances allow simplification and control of con-
founding variables. Simplification, however, may itself be an important shortcom-
ing in understanding inherently complex situations (4).
The laboratory approach has not been generally applied to transfusion error
analysis, although the announced introduction of simulated benign errors into
transfusion operations has been an effective means for increasing error detection.
Taswell et al. (14) demonstrated that by modifying work to demand staff attention
in looking for known introduced errors and by providing positive feedback when
they were found, the blood bank not only achieved an increased detection of the
introduced errors, but also increased the detection of real, previously undetected
errors from 4 in the first 3 months to 73 in the final 3 months of the study.
E. Event Reporting
A fifth approach to compiling information for the study of error is the event
report, including self-reporting as exemplified by the Aviation Safety Reporting
Managing Error for System Improvement 91
(22), which represents a continuum from the rare visible accident to the much
more frequent near misses. Linden et al.s (13) data from New York State can
illustrate this relationship with the iceberg in Figure 1. The fatalities from
hemolytic reactions due to ABO-incompatible blood transfusions are the visible
tip of the iceberg, the number of adverse reactions reported as being just below
the waterline, the number of incorrect units transfused by nurses as next, and the
unreported near misses as unknown.
A fundamental aspect of any event-reporting system is the identification or
detection of events that occur within an organization. Zapt and Reason (23)
indicate that error detection is the first step in error management. If an error
is not detected, it cannot be managed. They go on to point out that, from an
organizational point of view, it is very important that the error detection rate
is high; errors that are not detected for a long time could have disastrous
consequences. Thus, the goal of error management should be to increase error
detection or reporting rate. The number of events reported is an indicator of
an organizations detection sensitivity level (DSL). High reporting rates indi-
cate a high DSL; few reported events indicate a low DSL. A low DSL might
be considered an indicator of an inadequate error-detection and reporting ap-
proach. While the DSL should remain high, the event severity level (ESL) of the
incidents detected should decrease over time as corrective actions are im-
plemented (24).
In order to achieve a high DSL, an organization must remove any im-
pediments to reporting an event. Confidential no-fault reporting is one of the
best ways to encourage event reporting. Since the purpose of the event-reporting
system is to learn about how systems are operating, there must be separation
between event reporting and employee performance assessment approaches.
Research literature indicates that organizational encouragement such as confiden-
tial no-fault reporting significantly increases error detection and reporting (24).
Table 1 is a review of the strengths and limitations of the five error analysis
methods previously discussed.
A. Causes of Errors
Reason (12) has identified two major categories of failures or errors that occur in
complex systems: active and latent. The distinction hinges on both who initiated
the failure and how long it took to have an adverse effect (25,26). Active failures
are committed by people in direct contact with the human/system interface, i.e.,
94 Kaplan and Battles
Figure 2 The relationship between latent and active failures and misadventures.
fusing blood products. We can moderate human fallibility, but we can never
eliminate it completely. Active errors are tied to human cognitive processes
and associated behaviors. Rasmussen (27,28) gives us a useful taxonomy for
identifying and classifying these different types of human behavior underlying
human errors:
Skill-based behavior refers to routine tasks requiring little or no conscious
attention during task execution.
Rule-based behavior refers to familiar procedures applied to frequent
decision-making situations.
Knowledge-based behavior refers to problem-solving activities such as
when one is confronted with new situations for which no readily avail-
able standard solutions exist.
1. Skill-Based Behavior
Most health professionals operate in a skill-based behavioral mode for all of the
routine tasks that are carried out, from drawing blood to the transfusion process
96 Kaplan and Battles
itself. These highly skilled activities become routine and can be executed without
conscious thought. It is like riding a bicycleonce you learn you can do it
automatically. Driving a car is another examplewe can drive while listening to
the radio or talking to a passenger. It is because these skills are so often used that
one can perform with a very high level of reliability. We operate almost as if
we were on automatic pilot, performing virtually without thinking about what
we are doing, at almost an unconscious level. However, there are opportuni-
ties for failures when one operates in the skill-based mode. If we are distracted
or something interrupts the smooth flow of a skill-based routine, a skill-based
failure can occurcalled a slip. An example of a slip is being distracted by
someone in the parking lot and inadvertently locking the keys in the car or
arriving home only to discover that you forgot to stop at the store and buy milk
on the way home. Slips are unintended errors caused by either not doing some
part of the routine, an error of omission, or doing something that one should not
have done, like a repeating step in the process twice, which would be a failure of
commission. Slips cannot effectively be remediated by retraining an individual.
It is a waste of time because the individual already knows how to perform the
task at a very high level of accuracy and retraining is often insulting and
ineffective. Counseling employees to be more careful is equally ineffective as a
means of remediation. However, slips can be prevented by redesign of equipment
or procedures so that it is harder to make a slip. For example, feedback mecha-
nisms can be designed into the process that give clues to the individual as soon
as they have made a slip. Job aids such as a template for reviewing donor records
that highlight omissions or inconsistencies can help to prevent slips in the
skill-based mode.
2. Rule-Based Behavior
Rule-based behavior occurs at the conscious level within the context of the
situation that exists. Rule-based behavior involves recognizing the situation, then
selecting the proper routine or protocol that is called for in a given situation.
For example, if we are driving and come to a stop sign, we must decide what
rules to apply in this situation. If it is a two-way stop, then there is a given set of
traffic rules to follow, but if it is a four-way stop then there are different rules of
the road that we must follow. Rule-based behavior is an if-then condition. Failure
in rule-based behavior can occur at the different stages in this conscious decision
and action process. These failures are often referred to as mistakes. A mistake can
occur under two conditions: selecting the wrong rule for a given situation, or
selecting the correct rule but carrying it out incorrectly. Rule-based failures can
occur when someone carries out a procedure that they are not qualified to
perform. Another type of rule-based failure is inadequately assessing or verifying
the situation, which can result in a mistake in selecting the correct rule. An ex-
Managing Error for System Improvement 97
3. Knowledge-Based Behavior
Knowledge-based behavior involves solving unique problems or selecting a
plan of action in a new or unfamiliar setting. Knowledge-based behavior most
often occurs with new employees. They do not have the depth of experience to
operate in the skill-based mode or to draw from past experience to select the
appropriate rule or protocol to carry out a task or to solve a problem. Learners
such as medical residents and other trainees often operate in the knowledge-based
mode because the number of unique or new situations for them is significant
compared to the experienced individual or expert. Experienced individuals only
rarely operate in the knowledge-based mode. Thus, the expert and the novice are
likely to make different types of errors. The expert is most likely to make a slip
or an occasional rule-based error, while the novice is most likely to have
knowledge-based failures. If is possible for the expert to encounter unique
conditions and be placed in a situation where they can be subject to knowledge-
based failures. The procedures, equipment, and situation are all slightly different,
and their set of skills and rules may be inadequate for the new situation.
Examples of such conditions would be expert pilots moving from one type of
aircraft to another. The skills and rules used in operating a Boeing 737 are not the
same as for a Boeing 757. While it may not take an expert long to become
familiar with a new setting, there is a need for orientation and knowledge
transfer from the previous setting to the new one. This is why it is good practice
to have individuals recertified or credentialed when moving to a new job or
assuming new responsibilities.
Figure 3 summarizes the three levels of human behavior associated with
such common activity as driving a car.
98 Kaplan and Battles
C. Latent Failures
While we may never totally eliminate human or active errors, we can eliminate
the technical or organizational aspects that might set the person up for an active
failure. Latent or system failures include both technical and organizational
aspects. The technical aspects associated with latent failure include such things
as the design of equipment and software, the construction of facilities, including
mobiles, and materials. One aspect of organizational failures stems from normal
management considerations including the structure of the organization, planning
and scheduling, forecasting, budgeting, and allocating resources. The policies and
procedures in place in an organization can also be a source of latent failure, as
are the orientation, training, and selection of employees. The informal but very
real culture of an organization can be another source of latent failure. These latent
failures have the potential of setting up the individuals for failure. The adverse
safety consequences of normal technical and organizational decisions may lie
dormant for a very long time. Reason (12) has referred to latent error as
organizational pathogens, which wait to combine with the right active human
failure to have an adverse consequence.
It is difficult to recognize a latent error before the fact and to predict how
it will present itself in the future, since there are so many possible outcomes of
technical and management decisions. However, since an event represents a fixed
Managing Error for System Improvement 99
outcome, one is often able to identify the responsible latent error by reasoning
backward from the event. Once latent failures are recognized, they can be
diagnosed and corrected before they combine with an active error to produce a
bad outcome.
on finding the guilty party(s) who were negligent and committed the error.
The system searches for those negligent or less than perfect health professionals
to eliminate them and, in so doing, reduce error from the system. Human resource
policies encourage or mandate recording the number of errors or adverse events
within an employees personnel record. Such policies can lead to denial that errors
have occurred. If an error is caught and is not revealed and if no harm was caused
to the patient, then it is rationalized that it did not matter and need not be revealed.
Both conditions tend to result in underreporting of events. Despite the illusion of
perfect performance, Paget (31) points out that health care is error-ridden, inexact,
uncertain, and is practiced on the human body.
Reason (25) points out that the blaming individuals leads to ineffective
countermeasures: disciplinary action, exhortations to be more careful, retraining,
and writing new procedures to proscribe those actions implicated in some recent
event. He goes on to point out that these measures can have an impact at the outset
of some necessary safety program, but they have little or no value when applied
to a well-qualified and highly motivated work force. Figure 4 illustrates the issue
of assigning blame.
Rule-based
Qualifications HRQ Incorrect fit between an individuals qualification, training, or education and a particular task
Coordination HRC A lack of task coordination within a health-care team in an organization
Verification HRV Failures in the correct and complete assessment of a situation including relevant conditions
of the patient and materials to be used before starting the intervention
Intervention HRI Failures that result from faulty task planning (selecting the wrong protocol) and/or
execution (selecting the correct protocol but carrying it out incorrectly)
Monitoring HRM Failures during monitoring of process or patient status during or postintervention
Skill-based
Slips HSS Failures in performance of fine motor skills
Tripping HST Failures in whole body movements
3. Other
Managing Error for System Improvement
A. Patient-related factor PRF Failures related to patient characteristics or conditions beyond the control of staff and
influence treatment
B. Unclassifiable X Failures that cannot be classified in any other category
103
104
Kaplan and Battles
VI. SUMMARY
The safety of transfusion can be improved if one can identify errors that are an
indication of a systems weak points before they result in an adverse outcome to
a donor or a patient. Doing this requires a focus not only on adverse events, but
on the capture and recording of near-miss events as well. In order to capture
near-miss events, it is necessary for everyone in an organization to identify and
report those conditions and actions that have the potential to adversely affect
patient safety. It is also necessary to avoid assigning blame when an error is
identified, but rather to find the root causes of the error. Without an adequate
understanding of the causes of error, there is little likelihood the error can be
corrected and prevented in the future. It is essential to look for and to eliminate
those things that set up humans for failure. A useful goal is to create a safety
culture where everyone seeks to identify and report conditions that may compro-
mise transfusion safety.
106
Kaplan and Battles
REFERENCES
1. Linden JV, Kaplan HS. Transfusion errors: causes and effects. Transfus Med Rev
1994; 8(3):169183.
2. Guidelines for Good Manufacturing Practices. U.S. Food and Drug Administration,
1993.
3. Monger P. Packaging security in the pharmaceutical industry: an MCA inspectors
viewpoint. Pharmaceut Technol 1993; 4:81.
4. Nagel D. Human error in aviation operations. In: Wiener EL, ed. Human Factors in
Aviation. San Diego, CA: Academic Press, 1988:
5. Shulman IA, Lohr K, Derdiarian AK, Picukaric JM. Monitoring transfusion practices a
strategy for improving transfusion practices. J Bone Joint Surg 1992; 74-A:652658.
6. Honig CL, Bove JR. Transfusion-associated fatalities: review of Bureau of Biologics
reports 19761978. Transfusion 1980; 20:653661.
7. Myhre B. Fatalities from blood transfusion. JAMA 1980; 244:13331335.
8. Camp FR, Monaghan WP. Fatal blood transfusion reactions: an analysis. Am J
Forensic Med Pathol 1981; 2:143150.
9. Edinger S. A closer look at fatal transfusion reactions. Med Lab Obs 1985; 4:4145.
10. Sazama K. Reports of 355 transfusion-associated deaths: 1976 through 1985. Trans-
fusion 1990; 30:583590.
11. Mummert TB, Tourault MA. Review of transfusion related fatalities: many prevent-
able. Hosp Technol Scanner 1993; 4:13.
12. Reason J. Human Error. New York: Cambridge University Press, 1990.
13. Linden JV, Paul B, Dressler KP. A report of 104 transfusion errors in New York State.
Transfusion 1992; 32:601660.
14. Taswell HF, Smith AM, Sweatt MA, Pfaff KJ. Quality control in the blood bank: a
new approach. Am J Clin Pathol 1974; 62:491495.
15. Classen DC, Pestotnik SL, Evans RS, Burke JP. Computerized surveillance of adverse
drug events in hospitalized patients. JAMA 1991; 266:28172851.
16. Classen DC, Pestotnik MS, Evans RS, Lloyd JF, Burke JP. Adverse drug events in
hospitalized patients. JAMA 1997; 277:301306.
17. Reynard WD, Billings CE, Cheaney ES, Hardy R. The Development of the NASA
Aviation Safety Reporting System. Moffett Field, CA: National Aeronautics and
Space Administration Science and Technology Branch, 1986.
18. Billings, CE. Personal communication
19. Murphy WG, McClelland DBL. Deceptively low morbidity from failure to practice
safe blood transfusion: an analysis of serious blood transfusion errors. Vox Sang
1989; 57:5962.
20. Gambino R, Mallon P. Near misses: an untapped data base to find root causes. Lab
Rep 1991; 13:4144.
21. Van der Schaaf TW. Near miss reporting in the chemical process industry. Ph.D.
thesis, Eindhoven University of Technology, 1992.
22. Heinrich HW. Industrial Accident Prevention. New York: McGraw-Hill, 1931.
23. Zapt D, Reason JT. Introduction to error handling. Appl Psychol 1994; 43:427432.
24. Kaplan HS, Battles JB, Van der Schaaf TW, Shea CE, Mercer SQ. Identification and
108 Kaplan and Battles
I. INTRODUCTION
The clinical course of allogeneic bone marrow and peripheral blood stem cell
transplant patients is frequently complicated by graft-versus-host disease (GVHD).
This condition results when lymphocytes in the donor hematopoietic stem cell
(HSC) component recognize the HLA antigens of the recipient as foreign,
generating a characteristic immune response (1,2). Fever, diarrhea, liver function
test (LFT) abnormalities, and a characteristic cutaneous rash are the major clinical
manifestations of this condition. However, GVHD may also result from the
infusion of viable T lymphocytes within cellular blood components. This condi-
tion, which is accompanied by marrow aplasia and pancytopenia, was first
described in 1966 (3). In the following decades, subsequent reports established
transfusion-associated graft-versus-host disease (TA-GVHD) as a distinct disease
entity. TA-GVHD is resistant to most immunosuppressive therapeutic modalities.
Consequently, treatment is rarely successful, making the recognition of patient
groups warranting preventive measures essential.
Recently, new therapeutic strategies utilizing the potential graft-versus-
leukemia (GVL) effect of allogeneic lymphocytes have been developed, empha-
sizing the need to further understand the pathogenesis of TA-GVHD. Adoptive
immunotherapy, the infusion of allogeneic lymphocytes in patients who have
relapsed following allogeneic hematopoietic progenitor cell (HPC) transplanta-
tion, has been shown to produce both remissions and a clinical syndrome similar
to TA-GVHD (4). Whether the GVL effect is separable from GVHD is the object
of ongoing laboratory and clinical studies.
109
110 Webb and Anderson
IV. INCIDENCE
The epidemiological data on TA-GVHD are derived from analysis of reports of
single or very small groups of patients. A prospective study on the development
of TA-GVHD has never been undertaken and would be very difficult to perform.
Estimates of the incidence and identification of patient groups at risk are therefore
Graft-Versus-Host Disease 111
Table 4 TA-GVHD in
Immunocompetent Patients:
Clinical Setting
Pregnancy
Cholecystectomy
Cardiac surgery
Vascular surgery
Gastrointestinal surgery
Abdominal surgery
Alpha thalassemia
Liver transplantation
Pancreosplenic transplantation
114 Webb and Anderson
VI. PATHOGENESIS
The immune status of the host and the extent of HLA mismatch between donor
and recipient together determine the extent of any host-versus-graft response.
The ability of the transfusion recipient to mount an immune response against
donor T lymphocytes is fundamental to the pathogenesis of TA-GVHD. Thus,
Billingham (33) has proposed three requirements for the development of GVHD:
(a) differences in histocompatibility (HLA) antigens between the donor and the
recipient, (b) the presence of immunocompetent cells in the graft, and (c) inability
of the host to reject these immunocompetent cells. Usually the larger number of
immune cells in the immunocompetent host will eliminate the donor-derived
T cells via a host-versus-graft reaction. However, if an immunocompetent HLA-
heterozygous individual is transfused with even a small number of functional
T lymphocytes derived from a donor who is homozygous for one of the recip-
ients HLA haplotypes, the recipients immune system does not recognize the
major histocompatibility antigens on the donor cells as being foreign and is
therefore incapable of eliminating them. Some of the donor-derived T cells will
recognize those host HLA antigens that are encoded by the unshared haplotype
as being foreign, undergo clonal expansion, and establish TA-GVHD (26).
The TA-GVHD expressed under these circumstances may be expected to occur
regardless of the hosts immune status since the failure to eliminate donor-derived
T lymphocytes is based on the genetics of the HLA system rather than on
variables that contribute to immune competence per se.
Fast et al. (34) have provided important insights into the pathogenesis of
TA-GVHD, implicating recipient CD4+, CD8+, and natural killer cells in control-
ling TA-GVHD. In a mouse model, varying numbers of parental lymphoid cells
were injected into unirradiated F1 hybrid recipients, providing donor lymphocytes
homozygous for an HLA haplotype present in the recipient. The effect of selective
depletion of recipient CD4+, CD8+, and natural killer cells on the regulation of
Graft-Versus-Host Disease 115
TA-GVHD was assessed. Depletion of CD4+ cells increased the number of donor
cells necessary to induce TA-GVHD, while depletion of recipient CD8+ cells or
natural killer cells decreased the number of door cells required to produce
TA-GVHD. Thus, CD4+ cells may be involved in the pathogenesis of TA-GVHD,
and CD8+ and natural killer cells may be protective. According to this model,
patients at high risk for TA-GVHD would be those with impaired CD8 and natural
killer cell function, especially if they receive blood products with a one-way
HLA match.
No cases of TA-GVHD have been reported in individuals with AIDS,
further emphasizing the role of CD4+ and CD8+ lymphocytes in the pathogenesis
of TA-GVHD. It is possible that TA-GVHD may account for some of the
nonspecific signs and symptoms presently attributed to infections, drug reactions,
and other coexistent medical conditions in AIDS patients. Alternatively, it may
be that some qualitative aspect of the human immunodeficiency virus (HIV)
related immune deficit may alter the predisposition of AIDS patients to develop
TA-GVHD. The murine study reported by Fast et al. (34) suggests that the latter
explanation may be valid, as CD4+ lymphocyte function declines early while
CD8+ function is preserved until late in the course of HIV disease. In the mouse
model, both of these features would be expected to decrease the likelihood of
developing TA-GVHD. In contrast, it has been proposed that reactive recipient
CD8+ T cells are required for the development of GVHD (35). Activated
HIV-1infected CD4+ T cells express an HLA class 2derived peptide mimicked
by the carboxy-terminus of the HIV-1 envelope. It is hypothesized that the
activation of CD8+ T cells against the HIV-infected CD4+ T cells may preclude
the development of GVHD. Consequently, clarification of the role of both CD4+
and CD8+ cells in AIDS will likely provide further insight into the pathogenesis
of GVHD and vice versa.
In recipients of allogeneic HPC transplants, the spectrum between GVHD
and HLA alloimmunization and the extent of mononuclear cell microchimerism
depend on the dose of allogeneic cells transfused, the immune competence of the
recipient, as well as the extent of HLA similarity between donor and recipient
(36). For example, in an animal model of postbone marrow transplant GVHD,
reducing the number of cells transplanted resulted in a stepwise increase in
rejection rates of donor cells; conversely, reduction in the size of the marrow
innoculum could be compensated by increasing host immunosuppression (37).
Since transfused cellular blood products are rarely HLA-antigen tested or
matched, the first of Billinghams three requirements (33)that there be dif-
ferences in HLA between the donor and recipientis almost always present
in the setting of blood component transfusion. TA-GVHD is mediated by the
viable T lymphocytes that inevitably contaminate nonirradiated transfused cellu-
lar blood components. The naturally or iatrogenically immunosuppressed recipi-
ent has only a limited capacity to generate an effective host-versus-graft reaction,
116 Webb and Anderson
and greater HLA disparity increases the probability that donor lymphocytes will
attack host tissues.
VII. DIAGNOSIS
The differential diagnosis for TA-GVHD is broad: myriad factors such as infec-
tions and drug reactions can result in the development of fevers, skin rashes, and
LFT abnormalities. Histological findings in the skin and gastrointestinal tract may
suggest the diagnosis of TA-GVHD, but are not pathognomonic. The only
definitive approach to the diagnosis of TA-GVHD is the identification of donor-
derived lymphocytes in the circulation or tissues of the affected host (Table 5).
This requires either careful HLA typing or some other technique that reliably
distinguishes between host and donor cells (3841). Blood samples adequate for
HLA typing are frequently not available, since circulating host blood cells are
rapidly eliminated. Polymerase chain reactionbased methods for HLA typing
may be a useful substitute (4043). Otherwise, the patients HLA type may be
deduced from those of surviving first-degree relatives (26). Cytogenetics have
been employed in the event that donor and recipient have been of different gender
or when the disease has followed transfusion of granulocytes donated by individ-
uals with Philadelphia chromosome-positive chronic myeloid leukemia (44).
Increasingly sophisticated techniques for confirming the diagnosis of TA-GVHD
have included the detection of polymorphisms for restriction fragment lengths
and human microsatellite markers (27,41). Even so, this syndrome is still fre-
quently diagnosed only at autopsy.
VIII. THERAPY
Treatment of TA-GVHD is only rarely effective. Attempted immunosuppressive
therapies have included glucocorticoids, antithymocyte globulin, cyclosporine,
cyclophosphamide, and anti-T-cell monoclonal antibodies (8,23,25,4547). Al-
though some of these agents have been successful in the treatment of post-HPC
transplant GVHD, they have been ineffective for TA-GVHD. Rare responses to
some of the commonly used agents have been reported (7,48), contributing to
anecdotal experiences from which it is difficult to extract guidelines for clinical
practice. A therapeutic trial of glucocorticoids or other immunosuppressive agents
is often attempted but is frequently ineffective in this serious and increasingly
frequent disease. There appears to be no advantage to early diagnosis and
treatment, as patients diagnosed early in the course of the disease fare no better
than those diagnosed later. Thus, prevention is critically important.
have also been raised (49,50). However, irradiation is likely indicated within
genetically homogeneous populations, since transfusion of unirradiated compo-
nents between unrelated donor-recipient pairs may be expected to result in
TA-GVHD when a one-way HLA match occurs by chance.
The American Association of Blood Banks (AABB) currently recommends
the irradiation of blood components at 2.5 Gy to the center of the component,
with no area receiving less than 1.5 Gy (51). This irradiation dose is in excess of
the levels found to abrogate mixed lymphocyte culture (MLC) reactivity, as this
dose is inadequate to prevent TA-GVHD (52,53). Instead, the recommended
2.5 Gy dose is based on limiting dilution assays (LDAs) using visual assessment
of T-cell proliferation, which demonstrate a greater than 5 log reduction in viable
T cells. However, levels of host cytotoxic T-lymphocyte precursors (pCTLs) and
interleukin-2secreting helper T-lymphocyte precursors (pHTLs), which are not
measured in LDAs, may be more predictive of GVHD development following
allogeneic BMT (5458).
Significant variability in irradiation practice exists between centers. For
example, blood component irradiation practice was examined in a survey of 2250
blood centers, hospital blood banks, and transfusion services that are institutional
members of the AABB (59). Only 12.3% of the institutions had on-site facilities
for the irradiation of blood components. Of 9,397,516 components transfused in
1989, 952,516 (10.1%) were irradiated, and 44 cases of TA-GVHD were identi-
fied. There was marked variability in blood component irradiation practice, even
among groups in whom the risk of TA-GVHD was well defined. For example,
12, 19, and 32% of institutions did not provide irradiated components to recipients
of allogeneic BMT, patients receiving autologous BMT, and those with congenital
immunodeficiencies, respectively. Irradiated blood components were provided by
51.4, 34, 32, and 20% of institutions to patients with leukemias, Hodgkins
disease, non-Hodgkins lymphoma, and solid tumors, respectively, and 24.5%
provided irradiated blood products to patients with AIDS.
X. NEW DIRECTIONS
A. Leukoreduction to Prevent TA-GVHD
In theory, leukoreduction of cellular blood components could be used to decrease
TA-GVHD. However, since neither the number nor quality of T cells that mediate
GVHD are presently defined, targets for leukoreduction to prevent TA-GVHD
cannot be established. Further, TA-GVHD has been reported in both immu-
nocompetent and immunodeficient recipients of transfusions leukoreduced by
filtration, suggesting that standard leukoreduction procedures cannot completely
prevent TA-GVHD (60,61). Newer technologies appear capable of decreasing the
number of contaminating leukocytes (62); whether the levels of leukoreduction
Graft-Versus-Host Disease 119
REFERENCES
1. Ferrara JL, Deeg HJ. Graft versus host disease. N Engl J Med 1991; 324:667764.
2. Antin JH, Ferrara JLM. Cytokine dysregulation and acute graft-versus-host disease.
Blood 1992; 80:29642968.
3. Hathaway WE, Brangle RW, Nelson TL, Roeckel IE. Aplastic anemia and alympho-
cytosis in an infant with hypogammaglobulinemia: Graft-versus-host reaction? J Ped
1966; 68:713722.
4. Kolb HJ, Schattenberg A, Goldman JM, Hertenstein B, Jacobsen N, Arcese W,
Ljungman P, Ferrant A, Verdonck L, Niederwieser D, van Rhee F, Mittermueller J,
120 Webb and Anderson
Influence of cell dose and graft-versus-host disease on rejection rates after allogeneic
transplantation. Blood 1992; 79:16121620.
38. Kunstmann E, Bocker T, Roewer L, Sauer H, Mempel W, Epplen JT. Diagnosis of
transfusion-associated graft-versus-host disease by genetic fingerprinting and poly-
merase chain reaction. Transfusion 1992; 32:766770.
39. Suzuki K, Akiyama H, Takamoto S, Maruyama Y, Sakamaki H, Akagi K, Maeda Y,
Takenaka M, Onozawa Y. Transfusion-associated graft-versus-host disease in a
presumably immunocompetent patient after transfusion of stored packed red cells.
Transfusion 1992; 32:358360.
40. Hayakawa S, Chishima F, Sakata H, Fuji K, Ohtani K, Kurashina K, Hayakawa J,
Suzuki K, Nakabayashi H, Esumi M, Nemoto N, Sakurai I, Satoh K. A rapid
molecular diagnosis of posttransfusion graft-versus-host disease by polymerase chain
reaction. Transfusion 1993; 33:413417.
41. Wang L, Juji T, Tokunaga K, Takahashi K, Kuwata S, Uchida S, Tadokoro K, Takai
K. Brief Report: Polymorphic microsatellite markers for the diagnosis of graft-
versus-host disease. N Engl J Med 1994; 330:398401.
42. Saito M, Takamatsu H, Nakao S, Shiobara S, Matsuda T, Tajika E, Nakamura T, Kaji
T, Yoshida T. Transfusion-associated graft-versus-host disease after surgery for
bladder cancer. Blood 1993; 82:326330.
43. Uchida S, Wang L, Yahagi Y, Tokunaga K, Tadokoro K, Juji T. Utility of fingernail
DNA for evaluation of chimerism after bone marrow transplantation and for diagnos-
tic testing for transfusion-associated graft-versus-host disease [letter]. Blood 1996;
87:40154016.
44. Matsushita H, Shibata Y, Fuse K, Kimura M, Iinuma K. Sex chromatin analysis of
lymphocytes invading host organs in transfusion-associated graft-versus-host disease.
Virchows Archiv B Cell Pathol 1988; 55:237239.
45. Arsura EL, Bertelle A, Minkowitz S, Cunningham J, Jr., Grob D. Transfusion-
associated-graft-versus-host disease in a presumed immunocompetent patient. Arch
Int Med 1988; 148:19411944.
46. Otsuka S, Kunieda K, Hirose H, Takeuchi H, Mizutani M, Nagaya G, Sato G, Kasuya
S, Matsutomo K, Noma A, Saji S. Fatal erythroderma (suspected graft-versus-host
disease) after cholecystectomy. Transfusion 1989; 29:544548.
47. Vogelsang GB. Transfusion-associated graft-versus-host disease in nonimmunocom-
promised hosts. Transfusion 1990; 30:101103.
48. Cohen D, Weinstein H, Mihm M, Yankee R. Nonfatal graft-versus-host disease
occurring after transfusion with leukocytes and platelets obtained from normal
donors. Blood 1979; 53:10531059.
49. Lind SE. Has the case for irradiating blood products been made? Am J Med 1985;
78:543544.
50. Perkins H. Should all blood from related donors be irradiated? Transfusion 1992;
32:302303.
51. Preparation of blood components. In: Klein H, ed. Standards for Blood Banks
and Transfusion Services. Bethesda, MD: American Association of Blood Banks,
1996:14.
52. Sproul AM, Chalmers EA, Mills KI, Burnett AK, Simpson E. Third party mediated
Graft-Versus-Host Disease 123
I. INTRODUCTION
Although more than three centuries have passed since the recognition that
transfusion can be associated with deadly complications, it is only in the last
1015 years that pulmonary injury has been generally appreciated as a possible
transfusion outcome. In fact, before the mid-1980s the only well-recognized
manifestations of pulmonary injury from transfusion were anaphylactic reactions
and circulatory overload. Unfortunately, the respiratory system can be compro-
mised by another, immunologically driven type of reaction. Such reactions
were originally designated by a variety of descriptive terms, including non-
cardiogenic pulmonary edema (1), allergic pulmonary edema (2), hypersensitivity
reaction (3), and leukoagglutinin transfusion reaction (4). Only in the last decade
has there been a broader appreciation that this type of lung injury has an
immunological basis.
125
126 Popovsky
III. COMPLICATIONS
TRALI differs from ARDS in several important ways. Unlike ARDS with its
attendant morbidity and mortality (death rate of approximately 4050%), approx-
imately 80% of patients with TRALI improve both clinically and physiologically
within 4896 hours of the original insult, provided there is prompt and vigorous
respiratory support (58) While in many ARDS patients the lung injury is
irreversible, in TRALI the pulmonary lesion is typically transient. The pO2 levels
Transfusion-Related Acute Lung Injury 127
B. Bacterial Contamination
Fever, hypotension, and vascular collapse are prominent features of bacterial
contamination (11). Respiratory distress is infrequently observed. Patients may
present with disseminated intravascular coagulation. The onset of symptoms is
within 12 hours of transfusion of cellular or plasma-containing blood compo-
nents. Although platelet concentrates or apheresis platelets are the most frequently
implicated components (12), red cell concentrates may be involved.
V. INCIDENCE
The incidence rate of TRALI is unknown. In one study from the mid-1980s, 1 in
5000 plasma-containing transfusions were associated with this reaction (6,8). This
study took place during a period in which blood banks in the United States were
converting to red cell concentrates containing significantly less plasma (reduced
from an average of 100 to approximately 4060 mL); therefore, one might assume
that the frequency has decreased. On the other hand, the number of reports in the
literature has increased dramatically. From 1951 when the syndrome was first
described until 1985, there were fewer than 40 case reports in the English
language literature (1531). Since then descriptions of at least 94 recipient
reactions have appeared in the literature (8,9,3248,52,53,6769), and the author
is aware of an additional 65 unpublished cases.
In a study by Clarke et al. (41), 46 (0.34%) of 14,602 transfusion were
associated with severe respiratory reactions to random donor platelets over a
2-year period in a single general hospital. Rare reactions were also seen in
recipients of red cells. The reactions are most frequently seen in patients with
hematological malignancies. The average age of the platelets at transfusion was
4.5 days, significantly greater than that of the controls.
Other investigators have failed to find an at-risk population, although
recurring cases have been rarely described (39). The male:female ratio among
reactors is approximately 1:1 Cases have been described in both very young and
elderly transfusion recipients (from one month to 87 years) (8,39). Most patients
had no history of transfusion reactions, and there are no common denominators,
disease associations, or underlying conditions that necessitated the transfusions.
A report from New Zealand found an incidence of 0.001% of ARDS
reactions from 1981 to 1987 from a total of 440,000 transfused blood components
(49). The nearly 20-fold difference in reported incidents in these studies may
reflect the level of awareness at a medical center, rather than changes in
incidence. At the Mayo Clinic, for instance, transfusions are often administered
under the supervision of blood bank staff who are trained to identify transfusion
reactions.
There is reason to believe that TRALI may be significantly underdiagnosed.
In a series of 40 patients with pulmonary edema in the operative setting,
Cooperman and Price (50) found that 50% of cases were attributed to circulatory
overload or an unknown cause. It is conceivable that some of these cases
represented TRALI. Culliford and colleagues (25) described a type of non-
cardiogenic pulmonary edema in three patients following cardiopulmonary by-
pass that is consistent with this diagnosis, but tests that might have supported it
were not performed.
In an analysis of transfusion-associated fatalities reported to the U.S. Food
and Drug Administration from 1976 to 1985, acute pulmonary injury was
Transfusion-Related Acute Lung Injury 129
implicated in 12.1% (31) of 256 cases (51). In this study, respiratory death from
acute-onset pulmonary edema was the third most common cause of death from
transfusion. This type of complication was more frequent than bacterial contam-
ination or anaphylaxis. The relative importance of TRALI may increase as other
important complications, specifically transfusion-transmitted hepatitis, recede as
a consequence of improved blood donor screening and testing.
Finally, in a recent study of hypotensive reactions associated with platelet-
containing products, 3 of 24 reactions were consistent with, but unrecognized
as TRALI (52). This report underscores two points: (a) that TRALI remains
underdiagnosed and (b) that it may be confused with other complications of
transfusion.
VII. MECHANISM
Although the precise mechanism of TRALI is unknown, there are sufficient clues
to assume that it is an immune-mediated event. Unlike most immunologically
triggered transfusion reactions, in TRALI pathological antibodies are typically of
donor, rather than recipient, origin (Table 2). Numerous reports have docu-
mented the presence of human leukocyte antigen (HLA)specific antibodies or
leukoagglutinins in the plasma of the donors of implicated blood components
(3,4,8,18,26,2935,3739,42).
Popovsky and Moore (8) found such antibodies in 89% of 36 cases in one
series. In about half of the cases studied, the HLA-A or HLA-B antibodies of the
implicated donor corresponded with one or more HLA epitopes of the recipient
130 Popovsky
Table 2 Immunological
Findings in TRALI
Donor plasma
HLA-specific antibodies
Granulocyte-specific antibodies
Leukoagglutinins
Recipient plasma
HLA-specific antibodies
Granulocyte-specific antibodies
Leukoagglutinins
(8,26). Goeken and colleagues (29) as well as others have confirmed these
findings (31). In other cases, neutrophil-specific antibodies (anti-NA2, anti-5b,
and anti-NB2) have been identified in the serum of implicated units (27,32,37).
These antibodies are usually found in the blood of multiparous donors. On the
other hand, in 5% of reported cases, similar specificities of antibodies are
found in the pretransfusion serum of the transfusion recipient (5,7,8). Finally,
in 515% of cases, no antibody has been identified in either the patient or
the donor.
The likely explanation of antibodies in the donor, rather than antibodies in
the recipient, as the causative agent is that the substrate with which the
leukocyte antibodies can reactnamely, the recipients entire circulating and
marginated pool of leukocyteis far larger than the quantity of donor leukocytes
present in a single transfused component. It appears that TRALI begins with
passive transfer of antibody from donor plasma to the recipient (5,7), which sets
off a chain of reactions.
Underscoring the primacy of the antibody-mediated theory are a handful of
case reports of TRALI associated with interdonor incompatibility. In a recent
report, a 67-year-old male with acute myelogenous leukemia received a pool of
four platelet concentrates and developed the signs and symptoms of classic
TRALI. In the ensuing workup, one of the platelet donors was found to have
anti-A2 and anti-A28 in the plasma. The recipient tissue type was negative for
both A2 and A28, and the plasma did not contain HLA granulocyte antibodies.
One of the other donors of the platelets used in the pool was found to be A28
positive (53).
What is the relationship of these antibodies to this reaction? Much of the
current understanding is gleaned from studies of ARDS. Although the mecha-
nisms involved in the development of ARDS are complex, there is considerable
evidence to support a major role for complement activation in neutrophil influx
into the lung, causing damage to the pulmonary microvasculature. When comple-
Transfusion-Related Acute Lung Injury 131
this lipid developing during routine storage of blood components and that it
primes polymorphonuclear oxidase. These investigators (48) found that at the
time blood components (e.g., whole blood, red blood cells, platelet concentrate)
become outdated, they contain a priming agent that enhance polymorphonuclear
NADPH-oxidase activity by 2.1 to 2.8 fold. As this study represents the first
description of an alternative, nonantibody-mediated model of TRALI, other
laboratories will need to confirm these findings.
In a retrospective study from the same laboratory reporting the work
described above, 10 consecutive patients suspected of TRALI were evaluated for
PMN priming activity (62). These investigators used as controls patients having
febrile or urticarial reactions. They found that postreaction sera from TRALI
patients demonstrated a significant increase in PMN priming activity (2.1-fold)
compared with the patients prereaction sera as well as that of the control group.
In all cases the priming activity from serum samples was inhibited by pretreat-
ment of the isolated PMNs with 400 M WEB 2170, an inhibitor of the
platelet-activating factor receptor. Silliman and colleagues found that PMN-
generating lipids have a significant effect on lung function in an isolated, perfused
rat lung model (63). When lipopolysaccharide formed at day 42 of red blood cell
storage was infused, significant changes in pulmonary artery pressure and lung
weight were observed.
One other mechanism that merits discussion involves cytokines. Numerous
laboratories have shown that cytokines play a central role in the modulation of
inflammatory immune responses. Several reports implicate cytokines, including
tumor necrosis factor (TNF) and interleukin-8 in the pathogenesis of IgG-
mediated hemolytic transfusion reactions (HTRS) (64,65). Of interest is the
observation that patients experiencing HTRs may have hypoxemic or hypercapnic
respiratory failure or both (66). It may be relevant that during HTRs the
concentrations of TNF increase and TNF has been implicated in the development
of septic ARDS (66). It has been suggested that the release of significant
quantities of TNF from degranulating neutrophils may contribute to the injury of
pulmonary capillary endothelium seen in ARDS. How these observations pre-
cisely relate to TRALI remains a matter of conjecture.
VIII. DIAGNOSIS
Because there is no diagnostic test or pathognomonic sign, TRALI remains a
diagnosis of exclusion. One must rule out other causes of respiratory distress and
pulmonary edema in the transfusion setting: myocardial infarction, circulatory
overload, and bacterial infection. Normal central venous and pulmonary wedge
pressures are consistent with TRALI. The demonstration of lymphocytotoxic,
HLA-or granulocyte-specific antibodies in donor or recipient serum is strongly
Transfusion-Related Acute Lung Injury 133
IX. TREATMENT
While the earlier reports of TRALI described a fulminant picture of respiratory
distress, it is clear that not all cases are associated with life-threatening compli-
cations. Respiratory support should be as intensive as dictated by the clinical
picture. In almost all cases oxygen supplementation is necessary, and if the
hypoxemia is severe, intubation and mechanical ventilation are important inter-
ventions (6,8). Once a diagnosis is seriously entertained, therapeutic measures
should be started promptly. Pressor agents may be useful in case of sustained
hypertension. Corticosteroids are probably of marginal value, and diuretics have
no role because the underlying pathology involves microvascular injury, rather
than fluid overload (36).
XI. PREVENTION
Strategies aimed at reducing the incidence of reactions are complicated by the
absence of a profile of those recipients at greatest risk and by the lack of a
diagnostic test. Any effort will suffer for lack of sensitivity or specificity.
Therefore, measures must focus on limiting exposure. As a consequence, there
is no consensus on the subject, and few blood collectors have taken specific
steps. However, some investigators have made recommendations that include the
following (5):
1. Donors who have been implicated in TRALI should be permanently
deferred or subsequent donations limited to the production of frozen-
deglycerolized or washed red blood cells. Such steps will prevent the
use of plasma from blood components prepared from these donors.
134 Popovsky
XII. CONCLUSION
TRALI is an important, life-threatening transfusion complication, the incidence
rate of which is unknown but is most likely underrecognized. There may be
several mechanisms that lead to a common pathway of microvascular insult and
alveolar injury. In most cases, this syndrome is reversible, particularly with rapid
diagnosis and treatment.
REFERENCES
1. Carilli AD, Ramanamurty MV, Chang YS, et al. Noncardiogenic pulmonary edema
following blood transfusion. Chest 1978; 74:310312.
2. Kernoff PBA, Durrant IJ, Rizza CR, Wright FW. Severe allergic pulmonary oedema
after plasma transfusion. Br J Haematol 1972; 23:777781.
3. Wolf CFW, Canale VC. Fatal pulmonary hypersensitivity reaction to HLA-incompati-
ble blood transfusion: report of a case and review of the literature. Transfusion 1976;
16:135140.
Transfusion-Related Acute Lung Injury 135
24. Dubois M, Lotze MT, Diamond WJ, et al. Pulmonary shunting during leuko-
agglutinin-induced non-cardiac pulmonary edema. JAMA 1980; 244:21862189.
25. Culliford AT, Thomas S, Spencer FC. Fulminating noncardiogenic pulmonary edema.
A newly recognized hazard during cardiac operations. J Thorac Cardiovasc Surg
1980; 80:868875.
26. Popovsky MA, Abel MD, Moore SM. Transfusion-related acute lung injury associ-
ated with passive transfer of antileukocyte antibodies. Am Rev Respir Dis 1983;
128:185189.
27. Yomtovian R, Kline W, Press C, et al. Severe pulmonary hypersensitivity associated
with passive transfusion of a neutrophil-specific antibody. Lancet 1984; 1:244246.
28. Reese EP Jr, McCullough JJ, Craddock PR. An adverse pulmonary reaction to
cryoprecipitate in a hemophiliac. Transfusion 1975; 15:583588.
29. Goeken NE, Schulak JA, Nghiem DD, et al. Transfusion in donor-specific blood
transfusion patients resulting from transfused maternal antibody. Transplantation
1984; 38:306307.
30. Dhainaut JF, Brossard Y, Dimercurio JP, et al. Oedeme pulmonaire lesionnel gravis-
sisme au cours du 20 echange plasmatique pour incompatibilite Rhesus. In: Hemo-
perfusion, Echanges Plasmatique en Reanimation. Paris: Expansion Scientifique
Francaise, 1981:291298.
31. Campbell DA, Swartz RD, Waskeritz JA, Turcotte JG. Leukoagglutination with
interstitial pulmonary edema (letter). Transplantation 1982; 34:300301.
32. VanBuren NL, Stroncek DF, Clay MA, et al. Transfusion-related acute lung in-
jury caused by an NB2 granulocyte-specific antibody in a patient with thrombotic
thrombocytopenic purpura. Transfusion 1990; 30:4245.
33. Eastlund T, McGrath PC, Britten A, Propp R. Fatal pulmonary transfusion reaction
to plasma containing donor HLA antibody. Vox Sang 1989; 56:6366.
34. Eastlund DT, McGrath PC, Burkart P. Platelet transfusion reaction associated with
interdonor HLA incompatibility. Vox Sang 1988; 55:157160.
35. OConnor JC, Strauss RG, Goeken NE, Knox LB. A near-fatal reaction during
granulocyte transfusion of a neonate. Transfusion 1988; 28:173176.
36. Levy GJ, Shabot MM, Hart ME, et al. Transfusion-associated noncardiogenic pulmo-
nary edema: Report of a case and a warning regarding treatment. Transfusion 1986;
26:278281.
37. Nordhagen R, Conradi M, Dromtorp SM. Pulmonary reaction associated with
transfusion of plasma containing anti-5b. Vox Sang 1986; 51:102107.
38. DeWolf AM, Van Den Berg BW, Hoffman HJ, Zundert AA. Pulmonary dysfunction
during one-lung ventilation caused by HLA-specific antibodies against leukocytes.
Anesth Analg 1987; 66:463467.
39. Leaman B, Anderson D, Walker I, Heddle N. Two episodes of non-cardiogenic
pulmonary edema in a patient caused by the same donors blood (abstr). Transfusion
1994; 34(suppl):26S.
40. Win N, Kaye T, Jones L, Knowles S. Transfusion-related acute lung injury (TRALI)
(abstr). Transfus Med 1995; 5(suppl 1):33.
41. Clarke G, Podlosky L, Petrie L, Boshkov L. Severe respiratory reactions to random
donor platelets: an incidence and nested case-control study (abstr). Blood 1994;
84(suppl 1):465a.
Transfusion-Related Acute Lung Injury 137
42. Lin M, Chen CC, Hong CC, et al. Transfusion-associated respiratory distress in
Taiwan. Vox Sang 1994; 67:372376.
43. Reissman P, Manny N, Shapira SC, et al. Transfusion-related adult respiratory distress
syndrome. Isr J Med Sci 1993; 29:303307.
44. Popovsky MA, Chaplin HC, Moore SB. Transfusion-related acute lung injury (letter).
Transfusion 1993; 33:444445.
45. Silliman C, Pitman J, Thurman G, Ambruso DR. Neutrophil (PMN) priming agents
develop in patients with transfusion-related acute lung injury (abstr). Blood 1992;
80:(suppl 1):261a.
46. Bux J, Hoch J, Bindl L, et al. Transfusionassoziierte acute Lungeninsuffizienz. Dtsch
Med Wochenschr 1994; 119:1924.
47. Gans ROB, Duurkens VAM, van Zundert AA, Hoorntje SJ. Transfusion-related acute
lung injury. Intensive Care Med 1988; 14:654657.
48. Silliman CC, Thurman GW, Ambruso DR. Stored blood components contain agents
that prime the neutrophil NADPH oxidase through the platelet-activating-factor
receptor. Vox Sang 1992; 53:133136.
49. Henderson RA. Acute transfusion reactions. NZ Med J 1990; 103:509511.
50. Cooperman LH, Price HL. Pulmonary edema in the operative and postoperative
period: a review of 40 cases. Ann Surg 1970; 172:833891.
51. Sazama K. Reports of 355 transfusion-associated deaths: 1976 through 1985. Trans-
fusion 1990; 14:654670.
52. Hume HA, Popovsky MA, Benson K, Glasman AB, et al. Hypotensive reactions: a
previously uncharacterized complication of platelet reaction? Transfusion. In press.
53. Virchis AE, Contreras M, Novarrete C, Jan-Mohamed R, et al. Transfusion-related
acute lung injury (TRALI) due to inter-donor incompatibility (abstr). Blood. In press.
54. Jacob JS, Craddock PR, Hammerschmidt DE, Moldow CF. Complement-induced
granulocyte aggregation: an unsuspected mechanism of disease. N Engl J Med 1980;
302:789794.
55. Hammerschmidt DE, Jacob HS. Adverse pulmonary reactions to transfusion. Adv
Intern Med 1982; 27:511530.
56. Craddock PR, Hammerschmidt DE, Moldow CF, et al. Granulocyte aggregation as a
manifestation of membrane interactions with complement: possible role in leukocyte
margination microvascular occlusion, and endothelial damage. Semin Hematol 1979;
16:140147.
57. Shasby DM, Van Benthuysem KM, Tate RM, et al. Granulocytes mediate acute
edematous lung injury in rabbits and in isolated rabbits lungs perfused with phorbol
myristate acetate: role of oxygen radicals. Am Rev Respir Dis 1982; 125:443-447.
58. Sznajder JI, Fraiman A, Hal JB, et al. Increased hydrogen peroxide in the expired
breath of patients with acute hypoxemic respiratory failure. Chest 1989; 96:606612.
59. Larsen GL, McCarthy K, Webster RO, et al. A differential effect of C5a and C5a des
Arg in the induction of pulmonary inflammation. Am J Pathol 1980; 100:179192.
60. Seeger W, Schneider U, Kreusler B, et al. Reproduction of transfusion-related acute
lung injury in an ex vivo lung model. Blood 1990; 76:1438444.
61. Westaby S. Mechanisms of membrane damage and surfactant depletion in acute lung
injury. Intensive Care Med 1986; 12:25.
62. Silliman CC, Paterson A, Dickey WO, Stroncek D, Popovsky MA, Caldwell SA,
138 Popovsky
Ambruso DR. The association of biologically active lipids with the development of
transfusion-related acute lung injury: a retrospective study. Transfusion. In press.
63. Silliman C, Allard J, Voelkel N, Ambruso D. Plasma and lipids from stored packed
red cells (PRBCs) cause acute lung injury in an animal model (abstr). Blood 1996.
64. Davenport RD, Polak TJ, Kunkel SL. White cell-associated procoagulant induced by
ABO incompatibility. Transfusion 1994; 34:943949.
65. Davenport RD, Streiter RM, Sandiford TJ, Kunkel SL. Interleukin-8 production in
red blood cell incompatibility. Blood 1990; 76:24392442.
66. Capon SM, Goldfinger D. Acute hemolytic transfusion reaction, a paradigm of the
systemic inflammatory response: new insights into pathophysiology and treatment.
Transfusion 1995; 35:513520.
67. Boshkov L, Silliman C, Clarke G, Dickey W, Ambruso D. Transfusion-related acute
lung injury (TRALI) following platelet transfusion (abstr). Blood 1995; 86:1403.
68. Lindgren L, Ylihankala A, Holme L, Koskimies S, et al. Transfusion-related acute
lung injury (TRALI) after fresh-frozen plasma in a patient with a coagulopathy. Acta
Anaesthes Scand 1996; 40:641644.
69. Bux J, Becker F, Seeger W, Kilpatrick D, et al. Transfusion-related acute lung injury
due to HLA-A2-specific antibodies in recipient and NB1-specific antibodies in donor
blood. Br J Haem 1996; 93:707713.
7
Clinical Effects of the
Immunomodulation Associated
with Allogeneic Blood Transfusions
Jos O. Bordin
Escola Paulista de Medicina, So Paulo, Brazil
Morris A. Blajchman
McMaster University and Hamilton Centre Canadian Red Cross
Society, Hamilton, Ontario, Canada
I. INTRODUCTION
Considerable data have accumulated over the past two decades suggesting that
allogeneic blood transfusions (ABTs) may be associated with immunomodulation
in recipients (1,2). Depending on patient category, this immunomodulatory effect
can be clinically detrimental or beneficial (Table 1). Thus, it has been suggested
that the ABT-associated immunomodulation might adversely affect the overall
clinical outcome in patients undergoing curative surgery for a variety of malig-
nant tumors by downregulating the recipients immune system permitting unreg-
ulated tumor cell growth (35). In addition, many observational studies have
implicated this ABT-associated immunomodulatory effect with an increased
prevalence of postoperative bacterial infection episodes after abdominal, open-
heart, and orthopedic surgery (69). These adverse clinical effects, however, have
yet to be proven to be associated with ABT. In contrast, there is clear evidence
that ABT-associated immunomodulation can be beneficial for selected categories
of patients, such as: increasing allograft survival in renal allograft recipients (10);
decreasing the recurrence rate in women with recurrent spontaneous abortion
(11); and possibly reducing the relapse rate in patients with Crohns disease (12).
139
140 Bordin and Blajchman
This chapter summarizes the currently available evidence relating to the clinical
effects of ABT-associated immunomodulation.
Aa Bb Cc
Relative risk of tumor recurrence compared Arm (1) 1.8; p = 0.04 Arm (1) 0.96; p = 0.96 Arm (1) 0.91; p = 0.53
with untransfused patients Arm (2) 2.1; p = 0.01 Arm (2) 7.01; p = 0.006
Cancer-specific survival rate (1) 68% (1) vs (2) Transfusedd = 69%
(2) 64% Not transfused = 81%
p = 0.60 p = 0.20 p < 0.001
PRBCs = Packed red blood cells; BC-PRBCs = buffy coatreduced PRBCs; LR-Allogeneic PRBCs = leukocyte-reduced (48 hours old) allogeneic PRBCs.
aFrom Ref. 17.
bFrom Ref. 18.
cFrom Ref. 19.
dPatients transfused with either LR-PRBCs or BC-PRBCs.
141
142 Bordin and Blajchman
2.1, p = 0.01) or autologous (RR = 1.8, p = 0.04) blood, compared to subjects that
had not been transfused (17). Since this study actually compared outcomes among
patients receiving autologous blood versus recipients of buffy coatreduced
allogeneic blood, it is possible that the degree of leukocyte removal resulting from
buffy coat reduction may have diminished the ABT-associated immunomod-
ulatory effect that might have been observed had unmodified allogeneic blood
been used. Buffy coatreduced allogeneic blood represents the removal of
approximately 80% of the leukocytes present in a unit of whole blood. Interest-
ingly, this level of leukocyte reduction has been shown to prevent donor-specific
T-cell responsiveness associated with nonbuffy coatreduced blood (20).
Thus it is possible that the widespread use in Western Europe of buffy-coat
reduced allogeneic cellular blood components may significantly lessen the
immunomodulatory risk of cellular blood component transfusions in Western
Europe compared to that occurring in allogeneically transfused patients in North
America (20).
The second RCT, from Munich, Germany, randomized 120 patients with
potentially curative colorectal carcinoma to receive either leukocyte-reduced
allogeneic blood or predeposited autologous blood. After a median follow-up of
22 months, tumor recurrence was detected in 28.9% of the patients transfused
with allogeneic blood compared with 16.7% of the subjects transfused with
autologous blood (p = 0.11). The authors also reported that the need for allo-
geneic blood was an independent predictor of cancer recurrence (RR = 7.01; 95%
Cl = 1.7727.75; p = 0.006) (18).
The third prospective RCT was also from The Netherlands and compared
recipients of leukocyte-reduced allogeneic packed red blood cells (PRBCs) with
buffy coatreduced allogeneic PRBC transfusions in patients also undergoing
curative surgery for colorectal cancer. This study did not detect a difference in
patient survival between the two transfusion arms. As in the Rotterdam study,
transfused patients who received either allogeneic or autologous blood had a
significantly lower 3-year survival than untransfused study subjects (69% vs.
81%, p < 0.001) (19). Interestingly, recurrence rates appeared not to be affected
by the need for transfusion (30% vs. 26% p = 0.22).
In 1993, the results of the available observational studies of patients with
colorectal carcinoma were subjected to meta-analysis by two groups of investi-
gators independently (4,5). The literature-based meta-analysis from the first team
of investigators reported that the RRs of cancer recurrence, cancer-associated
death, and death from any cause in subjects transfused with allogeneic blood were
1.80, 1.76, and 1.63, respectively (4). These authors concluded that their analysis
supported the hypothesis that perioperative ABT was associated with an increased
risk of cancer recurrence and death from this disease (4). The second team of
meta-analysts concluded that perioperative ABT increased the RR of cancer
recurrence by 37% (95% Cl = 2056%) (5).
Blood Transfusion and Immunomodulation 143
Aa Bb Cc Dd Ee Ff
PRBCs = packed red blood cells; LR-WB (stored) = leukocyte-reduced whole blood filtered after storage); Autologous BC-PRBCs = autologous buffy
coatreduced PRBCs; Allogeneic BC-PRBCs = allogeneic buffy coat-reduced PRBCs; NS = not significant.
aFrom Ref. 7.
bFrom Ref. 17.
cFrom Ref. 8.
dFrom Ref. 24.
eFrom Ref. 25.
Bordin and Blajchman
than patients who received 99.98% leukocyte-reduced allogeneic blood (23% vs.
2%; p < 0.01).
In another RCT, which had been designed to measure cancer-related
prognosis, no difference was detected in the prevalence of postoperative infection
in study patients who were transfused with allogeneic buffy coatreduced PRBCs
compared to those who received autologous whole blood (17). In this study,
however, transfused subjects who received either allogeneic or autologous red
blood cell transfusions had a higher prevalence of postoperative infection than
those who were not transfused (17).
In the RCT from Munich, in which study subjects were assigned to receive
either autologous PRBCs or buffy coatreduced PRBCs, a significantly higher
postoperative infection rate in patients transfused with buffy coatreduced
PRBCs was seen compared to that in subjects transfused with autologous PRBCs
(27% vs. 12% p = 0.036) (8). Using multivariate regression analysis to adjust for
other risk factors, the RR of postoperative infections in the allogeneic group
versus that in the autologous transfusion group was 2.84 (p = 0.047; 95% Cl =
1.027.98). The observed number of noninfectious complications was similar in
the two groups of patients. In contrast to that seen in the study of Busch et al.
(17), autologous blood recipients did not have a greater prevalence of postoper-
ative infectious complications than that seen in those who were not transfused (8).
A three-arm RCT including 914 cardiac surgery patients has been re-
ported from Leiden, The Netherlands (24). In this study, patients were assigned
randomly to receive allogeneic leukocyte-reduced buffy coatreduced PRBCs
filtered within 24 hours of collection, allogeneic leukocyte-reduced buffy coat
reduced PRBCs filtered after storage, or nonleukocyte-reduced buffy coatreduced
PRBCs. Patients who received allogeneic leukocyte-reduced buffy coatreduced
PRBCs showed a lower prevalence of both postoperative infection (22.7% vs.
17.3%; p < 0.05) and mortality rate than those transfused with nonleukocyte-
reduced buffy coatreduced PRBCs. This study, however, detected no difference
in mortality or in the prevalence of postoperative infection (16.7% vs. 17.7%)
between the two leukocyte-reduced groups of study subjects (24).
Another RCT randomly assigned 589 colorectal carcinoma patients to
receive either allogeneic poststorage leukocyte-reduced buffy coatreduced
PRBCs or allogeneic buffy coatreduced PRBCs (25). The results indicated that
patients transfused with allogeneic poststorage leukocyte-reduced buffy coat
reduced PRBCs had a significantly lower prevalance of postoperative bacterial
infections than subjects who received allogeneic buffy coatreduced PRBCs (0%
vs. 18.3%, p < 0.001) (25).
Recently, yet another large multicenter RCT randomly assigned 697
patients undergoing colorectal surgery to receive either allogeneic leukocyte-
reduced buffy coatreduced PRBCs or allogeneic buffy coatreduced PRBCs.
Leukocyte reduction was performed within 48 hours of blood collection. No
146 Bordin and Blajchman
be done because of heterogeneity of the data from the six RCTs (29). It may,
however, be possible to do a MAP. Such an international collaborative individual
patient-based meta-analysis might allow for a definitive conclusion to be reached
as to whether ABTs increase susceptibility to postoperative infection. Such a
study has been proposed recently (23).
develop cytotoxic antibodies and thus are at greater risk for earlier and more
severe allograft rejection (36).
Patients given leukocyte-reduced blood components such as washed RBC
concentrates or frozen-deglycerolized RBCs have been shown to have poorer
one-year cadaveric graft survival than recipients of whole blood or unmodified
RBC concentrates, indicating that the allogeneic leukocytes present in the donor
blood participate in the production of the ABT-associated beneficial effect (37).
Further studies are required to understand how ABTs induce their clinical effect
in renal transplantation; however, ABT is still considered an efficacious interven-
tion that can be useful in the management of some patients scheduled for kidney
transplantation. This treatment modality continues to be used in donor-directed
renal allograft transplantation.
produce Th2 cytokines. This could result in the failure to produce the Th1 subset
(43). The Th1 subset when activated produces interleukin-2 (IL-2), interferon-
(IFN-), tumor necrosis factor- (TNF-), and lymphotoxin (LT), but not IL-4 or
IL-5. On the other hand, the Th2 sub-set of helper cells produces IL-4, IL-5, IL-6,
and IL-10, but not IL-2, IFN-, TNF-, or LT. Th1 cells stimulate those cells
involved in the cellular immune response, while Th2 cells are associated with the
humoral immune response. The B7-1 protein delivers a costimulatory signal
through CD-28 and CTLA-4 T cell receptors, which regulate IL-2 secretion, while
the B7-2 protein provides a critical early costimulatory signal that results in T-cell
clonal proliferation (42). Impairment of one of the costimulatory pathway signals
thus could result in T-cell anergy (41,44).
ABTs have been shown to be associated with various alterations in in vitro
measurements of immunological responsiveness in recipients. The most com-
monly observed functional modification is a lowering in the helper: suppressor
(CD4:CD8) lymphocyte ratio. Such an abnormality has also been shown to occur
in patients with hemophilia A after treatment with factor VIII concentrates (45).
Recently, two prospective clinical trials have reported that the use of very
high-purity factor VIII concentrates retards the decline in CD4 counts over time
compared with intermediate purity factor VIII concentrates (4648). Moreover,
CD4 cell counts were shown to remain relatively stable in HIV-infected hemo-
philiacs treated exclusively with recombinant factor VIII for 3.5 years (49).
However, the use of high-purity factor VIII products was shown to neither retard
the development of AIDS nor decrease the risk of death in HIV-infected patients
with hemophilia (50). In addition to quantitative alterations of Tcell subsets,
functional abnormalities in lymphocytes have also been described in patients with
hemophilia. These include decreased proliferative responses to mitogens, de-
creased natural killer (NK) cell activity, diminished cell-mediated immunity,
hypergammaglobulinemia due to polyclonal B cell activation, T-cell activation,
and inadequate monocyte function (45,51). It has been suggested that some of
these functional immunological alterations could be prevented with the use of
very high-purity factor VIII concentrates (52).
Other functional immunological alterations associated with ABT include
suppression of lymphocyte blastogenesis, decreased antigen presentation, and
reduction in delayed-type hypersensitivity (53).
ucts (1). The Class ll HLA antigens present in transfused allogeneic leukocytes
appear to elicit an immune response by recipients T cells, but HLA compati-
bility between blood donor and recipient may result in the persistence of circu-
lating donor mononuclear cells in the recipient (54,55). In this context, it has
been demonstrated, using the polymerase chain reaction technique, that trans-
fused male leukocytes can persist for 16 days in female patients receiving
multiple ABTs (56). This prolonged survival of small numbers of transfused
allogeneic leukocytes within the recipients circulation, a phenomenon known
as mononuclear microchimerism, can downregulate the immune response of
the recipient, inducing tolerance for donor alloantigens and predisposing the
recipient to the development of a chronic transfusion-associated graft-versus-host
disease (54,55).
In vitro investigations have suggested that leukocytes lose their immuno-
genicity during blood storage. Consequently, it has been hypothesized that
the transfusion of allogeneic blood, stored for prolonged periods of time, could
result in transfusion-associated immunomodulation due to recipient T-cell an-
ergy (44).
The hypothesis that the immunomodulatory effect of ABT is related to the
presence of allogeneic leukocytes has also been supported by research involving
experimental animals. Data from such studies indicate that ABTs accelerate tumor
growth and enhance metastatic nodule formation in both inbred and outbred
experimental animals (57,58). In such studies, it has been shown that allogeneic-
ally transfused experimental animals (mice and rabbits) inoculated with synge-
neic tumor cells develop significantly higher numbers of pulmonary nodules than
animals given syngeneic blood (57,58). These studies show that animals receiving
allogeneic buffy-coat leukocytes develop significantly higher numbers of pulmo-
nary nodules than animals given either plasma or prestorage leukocyte-reduced
whole blood (58). Conceivably, the prestorage removal of allogeneic leukocytes
prevented the accumulation of the soluble biological mediators synthesized
and released by the donor allogeneic leukocytes during storage. It is possible that
such biologically active substances are involved in the ABT-associated im-
munomodulation induced. Relevantly, allogeneic leukocytes have been recog-
nized as the blood component responsible for the increased susceptibility to
gut-derived infection in a murine model (59).
Further clues about the mechanism of ABT-associated immunomodulation
have been provided by experimental data showing that the ABT-associated tumor
growthpromoting effect can be adoptively transferred. In these experiments,
naive animals that had received spleen cells from allogeneically transfused
animals (inbred and outbred) developed a greater number of pulmonary meta-
static nodules than was observed in animals that had received spleen cells from
animals, transfused with syngeneic blood. This ABT-associated tumor growth
promoting effect could not be adoptively transferred using spleen cells derived
152 Bordin and Blajchman
from animals that had been transfused with prestorage leukocyte-reduced al-
logeneic blood (57). Additionally, it has been observed that the ABT-associated
immunomodulatory effect occurred following the infusion of ABT-conditioned
splenic T cells but was not seen after the infusion of ABT-conditioned splenic B
cells, even though the presence of the B cells enhanced the extent of the tumor
growthpromoting effect of the ABT-conditioned splenic T cells.
The ABT-associated tumor growthpromoting effect also was observed in
adoptive-transfer experiments in which ABT-conditioned spleen cells were in-
stilled intraperitoneally in diffusion chambers that allowed only the release of
soluble substances. The results of these latter experiments suggest that the
ABT-associated tumor growthpromoting effect might be mediated by biologi-
cally active substances released by conditioned T cells. The nature of these
substances still needs to be defined.
The mechanism of the ABT-associated immunomodulatory effect thus is
still unresolved. It is probably due to a combination of mechanisms involving
active immunosuppression, host anergy and clonal deletion. With regard to clonal
deletion, the infusion of foreign MHC antigens could result in the deletion of
recipients T-cell clones, whose T-cell receptor is directed against foreign MHC
antigens. The development of an active suppressor cell network following ABT
thus may result in shift towards a Th2-type immune response. Preliminary clinical
data have recently suggested that ABTs elicit a Th2-type response predominantly
(60). Relevantly, a significant increase in soluble IL-2R and IL-6 concentration
has been detected in colorectal carcinoma patients receiving allogeneic whole
blood transfusions but not in patients transfused with leukocyte-reduced al-
logeneic blood (61).
Several in vitro studies have shown that low molecular weight com-
ponents found in factor VIII products inhibit the proliferative response of
mononuclear cells to phytohemaglgutinin (52). In these studies, high-purity
factor VIII concentrates have been shown to reduce the induced expression of
T-cell activation molecules such as the IL-2 receptor (CD25), the transferrin
receptor (CD71), CD38, CD11a/CD18, and HLA-DR (52). This inhibitory action
of factor VIII concentrates may, at least in part, be due to contamination with
transforming growth factor- (TGF-) (62). TGF- may also be involved in
mechanisms by which veto cells downregulate the immune responsiveness of
the host (63).
In contrast to active suppression, anergy refers to nonreactivity by the host.
As outlined earlier, the absence of costimulatory signals may result in T-cell
unresponsiveness. Of interest, it has been shown that IL-10, a Th2-type cytokine,
prevents in vitro expression of the CD80 family of costimulatory molecules, on
macrophages (64).
Blood Transfusion and Immunomodulation 153
VII. SUMMARY
The potential clinical importance of the immunomodulatory effects associated
with ABT is one of the major current concerns in transfusion medicine. Many
questions relating to this phenomenon remain to be elucidated. Data from both
retrospective and prospective studies, including three RCTs, addressing the issue
of immunomodulatory effects of perioperative ABT on cancer prognosis have
yielded contradictory results. The conclusions from six RCTs examining the
prevalence of postoperative septic complications following ABTs also have been
contradictory. In contrast, there is clear-cut evidence that ABT-associated im-
munomodulation might be beneficial for selected groups of patients. Accordingly,
transfusions of allogeneic cellular blood components have been shown to improve
renal allograft survival as well as reducing the recurrence rate in women with
recurrent spontaneous abortions.
The immunomodulatory effect of ABT is generally accepted as being
mediated by the allogeneic leukocytes and/or their products present in transfused
cellular allogeneic blood products. The presence of the allogeneic leukocytes in
these products has thus been associated with diverse adverse biological activities,
including febrile transfusion reactions, graft-versus-host disease, alloimmuniza-
tion to leukocyte antigens causing platelet refractoriness, and immunomodulation.
Data from animal-based research suggest that the ABT-associated im-
munomodulatory effect is immunologically mediated and that this effect is due
to the presence of allogeneic leukocytes in the transfused blood products. Accord-
ingly, the experimental animal data show that prestorage leukocyte reduction
might prevent the ABT-associated growth enhancement of animal tumors. A sim-
ilar protective effect of leukocyte reduction has also been observed in some
human studies of ABT-associated postoperative bacterial infections, but clear-cut
evidence for the clinical benefit of leukocyte reduction is not yet available.
REFERENCES
1. Bordin JO, Heddle NM, Blajchman MA. Biologic effects of white blood cells present
in transfused cellular blood products. Blood 1994; 84:17031721.
2. Meryman HT. Transfusion induced alloimmunization and immunosuppression and
the effect of white blood cell depletion. Transfus Med Rev 1989; 3:180193.
3. Francis DMA. Relationship between blood transfusion and tumour behaviour. Br J
Surg 1991; 78:14201428.
4. Chung M, Steinmetz OK, Gordon PH. Perioperative blood transfusion and outcome
after resection for colorectal carcinoma. Br J Surg 1993; 80:427432.
5. Vamvakas EC, Moore SB. Perioperative blood transfusion and colorectal cancer
recurrence: a qualitative statistical overview and meta-analysis. Transfusion 1993;
33:754765.
6. Vignali A, Braga M, Gianotti L, Radaelli G, Gentilini O, Russo A, Di Carlo V. A
single unit of transfused allogeneic blood increases postoperative infections. Vox
Sang 1996; 71:170175.
7. Jensen LS, Andersen AJ, Christiansen PM, et al. Postoperative infection and natural
killer cell function following blood transfusion in patients undergoing elective
colorectal surgery. Br J Surg 1992; 79:513516.
8. Heiss MM, Mempel W, Jauch KW, et al. Beneficial effect of autologous blood
transfusion on infectious complications after colorectal cancer surgery. Lancet 1993;
342:13281333.
9. Vamvakas EC, Moore SB. Blood transfusion and postoperative septic complications.
Transfusion 1994; 34:714727.
10. Opelz G. The role of HLA matching and blood transfusions in the cyclosporine era.
Transplant Proc 1989; 21:609612.
11. Coulam CB, Clark DA, Collins J, et al. Worldwide collaborative observational study
and meta-analysis on allogeneic white blood cell immunotherapy for recurrent
spontaneous abortion. Am J Reprod Immunol 1994; 32:5572.
12. Tartter PI, Heimann TM, Aufses, Jr, AH. Blood transfusion, skin test reactivity, and
lymphocytes in inflammatory bowel disease. Am J Surg 1986; 151:358361.
13. Gantt CL. Red blood cells for cancer patients. Lancet 1981; 2:363.
14. Bordin JO, Blajchman MA. Immunosuppressive effects of allogeneic blood transfu-
sions: Implications for the patient with a malignancy. Hematol Oncol Clin North Am
1995; 9:205218.
15. Vamvakas EC. Perioperative blood transfusion and cancer recurrence: meta-analysis
for explanation. Transfusion 1995; 35:760768.
16. Vamvakas EC. Transfusion-associated cancer recurrence and postoperative infection:
meta-analysis of randomized, controlled clinical trials. Transfusion 1996; 36:175186.
17. Busch ORC, Hop WCJ, van Papendrecht MAWH, Marquet RL, Jeekel J. Blood
transfusions and prognosis in colorectal cancer. N Engl J Med 1993; 328:1372 1376.
18. Heiss MM, Jauch KW, Delanoff C, Mempel W, Schildberg FW. Blood trans-
fusion modulated tumor recurrencea randomized study of autologous versus
allogeneic blood transfusion in colorectal cancer surgery. J Clin Oncol 1994;
12:18591867.
Blood Transfusion and Immunomodulation 155
19. Houbiers JGA, Brand A, van de Watering LMG, et al. Randomised controlled trial
comparing transfusion of leucocyte-reduced or buffy-coat-reduced blood in surgery
for colorectal cancer. Lancet 1994; 344:573578.
20. van Prooijen HC, Aarts-Riemens MI, van Oostendorf WR, Hene RJ, Gmelig-Meyling
FHJ, de Weger RA. Prevention of donor-specific T-cell unresponsiveness after
buffy-coat-reduced blood transfusion. Br J Haematol 1995; 91:219223.
21. Cerhan JR, Wallace RB, Folsom AR, Potter JD, Munger RG, Prineas RJ. Transfusion
history and cancer risk in older women. Ann Inter Med 1993; 119:815.
22. Dzik S, Blajchman MA, Blumberg N, Kirkley SA, Heal JM, Wood K. Current
research on the immunomodulatory effect of allogeneic blood transfusion. Vox Sang
1996; 70:187194.
23. Blajchman MA. Allogeneic blood transfusions, immunomodulation, and postopera-
tive bacterial infection: Do we have the answers yet? Transfusion 1997; 37:121125.
24. van de Watering LM, Houbiers JG, Hermans J, et al. White blood cell depletion
reduces postoperative mortality in patients undergoing cardiac surgery (abstract
#SY3DI-06). Proceedings of the 24th Congress of the International Society of Blood
Transfusion, Mukuhari, Japan, 1996, p. 37.
25. Jensen LS, Kissmeyer-Nielsen P, Wolff B, Qvist N. Randomised comparison of white
blood cell-reduced versus buffy-coat-poor transfusion and complications after col-
orectal surgery. Lancet 1996; 348:841845.
26. Houbiers JG, van de Velde CJ, van de Watering LM, Hermans J, Schreuder S, Bijnen
AB, Pahplatz P, Schattenkerk ME, Wobbes T, de Vries JE, Klementschitsch P, van de
Maas, AH, Brand A. Transfusion of red blood cells is associated with increased
incidence of bacterial infection after colorectal surgery: a prospective study. Transfu-
sion 1997; 37:126134.
27. Dawes LG, Aprahamian C, Condon RE, Malongi MA. The risk of infection after
colon injury. Surgery 1986; 100:796803.
28. Pinto V, Baldonedo R, Nicolas C, Barez A, Perez A, Aza J. Relationship of transfusion
and infectious complications after gastric carcinoma operations. Transfusion 1991;
31:114118.
29. Vamvakas EC, Blajchman MA. A proposal for an individual patient data-based
meta-analysis of randomized controlled trials of allogeneic transfusion and postoper-
ative bacterial infection. Transfus Med Rev 1997; 11:180194.
30. Opelz G, Sengar DPS, Mickey MR, Terasaki PI. Effect of blood transfusions on
subsequent kidney transplants. Transplant Proc 1973; 5:253259.
31. Opelz G, Terasaki PI. Improvement of kidney-graft survival with increased numbers
of blood transfusions. N Engl J Med 1978; 299:799803.
32. Blajchman MA, Singal DP. The role of red blood cell antigens, histocompatibility
antigens, and blood transfusions on renal allograft survival. Transfus Med Rev 1989;
3:171179.
33. Norman DJ, Wetzsteon P, Barry JM, Fischer S. Blood transfusions are beneficial in
HLAidentical sibling kidney transplants. Transplant Proc 1985; 17:23472348.
34. van Twuyver E, Mooijaart RJD, ten Berge IJM, et al. Pretransplantation blood
transfusion revisited. N Engl J Med 1991; 325:12101213.
156 Bordin and Blajchman
35. Ross WB, Yap PL: Blood transfusion and organ transplantation. Blood Rev 1990;
4:252258.
36. Sells RA, Scott MH, Prieto M, Bone JM, Evans GM, Millar R, Hillis AN. Early
rejection following donor-specific transfusion prior to HLA-mismatched living re-
lated renal transplantation. Transplant Proc 1989; 21:11731174.
37. Horimi T, Terasaki PI, Chia D, Sasaki B. Factors influencing the paradoxical effect
of transfusions on kidney transplants. Transplantation 1983; 35:320323.
38. Unander AM. The role of immunization treatment in preventing recurrent abortion.
Transfus Med Rev 1992; 6:116.
39. Tartter PI, Driefuss RM, Malon AM, Heimann TM, Aufses AH. Relationship of
postoperative septic complications and blood transfusions in patients with Crohns
disease. Am J Surg 1988; 155:4348.
40. Peters WR, Fry RD, Fleshman JW, Kodner IJ. Multiple blood transfusions reduce the
recurrence rate of Crohns disease. Dis Col Rect 1989; 32:749753.
41. Mincheff MS, Meryman HT. Costimulatory signals necessary for induction of T cell
proliferation. Transplantation 1990; 49:768772.
42. Guinan EV, Gribben JG, Boussiotis VA, Freeman GJ, Nadler LM. Pivotal role of the
B7:CD28 pathway in tranplantation tolerance and tumor immunity. Blood 1994;
84:32613282.
43. Webb LMC, Feldmann M. Critical role of CD28/B7 coestimulation in the develop-
ment of human Th2 cytokineproducing cells. Blood 1995; 86:34793486.
44. Mincheff MS, Meryman HT, Kapoor V, Alsop P, Wotzel M. Blood transfusion and
immunomodulation: a possible mechanism. Vox Sang 1993; 65:1824.
45. Watson HG, Ludlam CA. Immunological abnormalities in haemophiliacs. Blood Rev
1992; 6:2633.
46. de Biasi R, Rocino A, Miraglia E, Mastrullo L, Quirino AA. The impact of a very
high purity factor VIII concentrate on the immune system of human immuno-
deficiency virus-infected hemophiliacs: a randomized, prospective, two-year compar-
ison with an intermediate purity concentrate. Blood 1991; 78:19191922.
47. Seremetis SV, Aledort LM, Bergman GE, et al. Three-year randomised study of
high-purity or intermediate-purity factor VIII concentrates in symptom-free HIV
seropositive haemophiliacs: effects on immune status. Lancet 1993; 342:700 703.
48. Hilgartner MW, Buckley JD, Operskalski EA, Pike MC, Mosley JW. Purity of factor
VIII concentrates and serial CD4 counts. Lancet 1992; 341:13731374.
49. Mannucci PM, Brettler DB, Aledort LM, et al. Immune status of human im-
munodeficiency virus seropositive and seronegative hemophiliacs infused for 3.5
years with recombinant factor VIII. Blood 1994; 83:19581962.
50. Goedert JJ, Cohen AR, Kessler CM, et al. Risks of immunodeficiency, AIDS, and
death related to purity of factor VIII concentrate. Lancet 1994; 344:791792.
51. Blumberg N, Heal JM. Effects of transfusion on immune function: cancer recurrence
and infection. Arch Pathol Lab Med 1994; 118:371379.
52. Vermont-Desroches C, Rigal D, Blourde C, Bernaud J. Immunosuppressive property
of a very high purity antihaemophilic preparation: a low molecular weight component
inhibits an early step of PHA induce cell activation. Br J Haematol 1992; 80:370377.
53. Blajchman MA, Bordin JO. Mechanisms of transfusion-associated immunosuppres-
sion. Curr Opin Hematol 1994; 1:457461.
Blood Transfusion and Immunomodulation 157
Roger Y. Dodd
Holland Laboratory, American Red Cross, Rockville, Maryland
I. INTRODUCTION
Infection has always been an adverse outcome of blood transfusion. A great deal
of effort has been directed towards reducing the risk of this outcome, and these
efforts have been extraordinarily successful, so that it is true that, in objective
terms, the blood supply is safer than it has ever been (13). Nevertheless, the issue
of blood safety continues to attract a great deal of attention from the media,
politicians, and professionals. As a natural consequence of this attention, there
continues to be interest in further reduction of the frequency of transfusion-
transmitted infection. However, an important component of any program de-
signed to improve safety is an assessment of the current level of risk, along with
the benefits attributable to the proposed intervention. This should form a basis for
logical decision making and appropriate use of resources. In addition, it is
important that responsible and accurate information about risk be freely available
to those who prescribe or receive blood components. At the same time, it must
be recognized that public sentiment will not always support decisions made
purely upon the basis of costs and benefits. It is clear, for example, that there
is little tolerance for failure to take actions that may prevent transmission of
HIV, whereas there is much less concern about the greater risk of transfusion-
associated bacterial sepsis.
Transfusion-transmitted disease occurs because our measures to assure
blood safety are not perfect. At interview, donors may be unaware of their own
risk of infectivity or deny it. Laboratory errors may occur, and even if they do
not, current serological tests cannot identify all infectious individuals, since virus
159
160 Dodd
A. Surveillance
Surveillance is an organized approach to collecting and reviewing data about
posttransfusion disease or infection. It is an important tool but cannot necessarily
be used to generate useful estimates of risk. Two aspects of surveillance are
applicable. In the first, the blood provider or some other agency actively solicits
reports of posttransfusion disease from establishments that have used the blood.
This procedure requires a high degree of attention from the medical community
and effective record keeping across institutions. Even so, a reporting system will
only capture cases that are recognized, usually as a result of overt sickness,
although some cases come to light as a result of serological investigation. Thus,
even when successful in identifying disease, reporting is an insensitive measure
of the frequency of posttransfusion infection. Also, it suffers from the disadvan-
tage that there is not necessarily a causal linkage between transfusion and disease;
this is particularly true when, as is the case for many infections, incidence rates
in the community exceed those among blood recipients.
Formal programs of surveillance for posttransfusion disease have been
established at the national level in at least two countries. The hemovigilance
system in France involves a formal mechanism of reporting and assessing adverse
outcomes, as does the English SHOT (Serious Hazards of Transfusion) program.
These programs have been productive but suffer from the fact that there is really
no control over the initial recognition and reporting of posttransfusion disease.
Overview of Infectious Disease 161
In the United States, adverse effects of transfusion (with special focus on deaths)
must be reported to the Food and Drug Administration (FDA).
The second aspect of surveillance relates to follow-up investigation of
locally or nationally reportable disease. For example, in the United States,
viral hepatitis and acquired immunodeficiency syndrome (AIDS) are reportable.
In some states human immunodeficiency virus (HIV) infection is also reportable.
Further studies are performed to assess risk factors associated with these reported
diseases; for AIDS, all cases are investigated, whereas for hepatitis, only a
proportion are evaluated. It is reasonable to suppose that the majority of AIDS
cases are reported, but more intensive local surveillance clearly shows that
hepatitis is underreported (4,5). Again, surveillance mechanisms will only iden-
tify disease, and even in the case of HIV, where every infection will likely result
in disease, the length of the incubation period precludes contemporaneous esti-
mates of infection rates. Nevertheless, surveillance studies do demonstrate tem-
poral variation. They have effectively demonstrated a continuing reduction in the
incidence of hepatitis that is epidemiologically linked to transfusion (68) and
have revealed that, by December 1998, only 39 of the total of 8760 transfusion-
associated AIDS cases in the United States could be linked to transfusions given
after the implementation of anti-HIV testing (9). It should be noted that great care
is necessary in attributing infection to transfusion, and many cases of apparent
transfusion AIDS were not confirmed upon careful follow-up (10). In some cases,
surveillance data may be the only available way to estimate risk, as is the case
for transfusion-associated malaria in the United States.
B. Prospective Studies
Conceptually, the most satisfactory way to define the risk of transfusion-associated
infection is to perform a prospective study on a population of blood recipients,
thus providing a direct measure of the frequency of infection. This necessitates
the identification of a suitable recipient population, collection of pretransfusion
samples, and posttransfusion follow up with sampling over a sufficient time
period for evolution of markers of infection. The study population should be
selected to avoid confounding factors and with the expectation that the recipients
will survive for a reasonable amount of time after transfusion. A good example
of such a study is the series of investigations performed by Nelson and colleagues
on cardiac surgery patients in Baltimore and Houston (1114). Much greater
value is gained by retaining samples of all donations along with sufficient
information to access the donors again in the future. In the context of posttransfu-
sion follow-up studies, the classic examples are the hepatitis studies performed
in the late 1970s, particularly in the United States (1518). Such studies have
proven to be a rich mine of information over many years, although it should be
emphasized that great caution must be taken in extrapolating all of the data to
162 Dodd
current times. However, Alters study (17) continues to the present day. Even
further value may be obtained from prospective studies that include a controlled
evaluation of an intervention. For example, the posttransfusion study reported by
Blajchman and colleagues assessed the impact of surrogate markers [alanine
aminotransferase (ALT) and hepatitis B core antibody (anti-HBc)] in reducing the
incidence of posttransfusion hepatitis (19). Similarly, studies by Bowden and
colleagues compared the frequency of cytomegalovirus (CMV) infection and
disease in bone marrow transplant patients receiving CMV-seronegative or leuko-
reduced blood components (20).
The major advantage of a properly conducted prospective study is that each
posttransfusion infection may be unequivocally identified and that proper atten-
tion to follow-up testing of the donors can clearly establish that the infection was
indeed acquired by transfusion of a given donors blood. Ideally, such investiga-
tion should include serological or genomic characterization of the specific strain
of the isolates from donor and recipient. The size of the recipient population is
known, along with the number of products transfused, so infection rates can be
defined, along with confidence intervals.
The major disadvantages of prospective studies are their complexity and
cost. In fact, as will be shown below, the frequency of residual infections is now
such that enormous studies have to be performed even to identify a single
infection. Nelsons studies, for example, included up to 9294 transfused, and 2238
untransfused patients, involving transfusion of 120,312 components (13). Even if
one or two infections are identified, the confidence intervals for the resulting risk
estimates are large. More useful data on the risk of hepatitis C were, however,
obtained from this study, with the earliest report showing transmission of HCV
from 0.45% of donations before testing was initiated and 0.19 and 0.03% risks
after the respective initiation of surrogate testing and version 1.0 anti-HCV tests
(12). The resources needed to mount comprehensive prospective studies are no
longer available, and in any case it could be argued that the expenditure would
not be justified. Another difficulty is the extent to which a study can be regarded
as representative. As pointed out above, the posttransfusion hepatitis studies of
the 1970s, although still widely quoted, cannot be regarded as representative of
todays situation, as they involved very different donor populations (some of which
were paid) and less comprehensive procedures for donor screening and collection
of medical histories. Nelsons study was performed in two metropolitan areas that
could not be presumed to be representative of the country as a whole. Finally, the
tests involved may not be adequate to provide the best measure; the ALT tests
used in the original post transfusion hepatitis (PTH) studies were neither specific
nor sensitive, and reevaluation of Nelsons study on HCV seroconversion, using
a newer test procedure, essentially doubled the per-unit incidence estimate (12,14).
Another direct measure of the risk of infectivity for HIV was developed by
Busch et al. (21) and Vyas et al. (22) who pooled samples of blood donations and
Overview of Infectious Disease 163
used viral culture and PCR technology to look directly for evidence of HIV in
blood, tested and issued as seronegative. Ultimately, this study yielded one isolate
from an investigation representing the equivalent of 160,000 donations (21,22).
Clearly, this represented an enormous effort, which was potentially subject to
criticism about the sensitivity or specificity of the methods involved. In addition,
this heroic study generated but a single data point, with its resultant wide
confidence interval.
Finally, in cases where a diagnostic test is available, but is not used for
donor screening, it is possible to perform a seroprevalence study among blood
donors. If there is some knowledge about the frequency of transmission of the
agent from seropositive donors, then it is possible to make a reasonable estimate
of the risk of recipient exposure and infection in the absence of the test. Once a
test is implemented, however, the problem becomes the measurement of efficacy
of the test and other methods must be used. At this time, donor seroprevalence
studies of this type are being performed in order to assess the risk of exposure to
Trypanosoma cruzi, the agent of Chagas disease, in the United States (23,23a).
States detects seroconversion some 23 days earlier than those in use in 1990 (34).
These data have been used to arrive at an estimate of risk of 1:340,000 per
component in the United States as of the beginning of 1995. This estimate is
reduced to 1:420,000 if an adjustment is made for the impact of other tests (28).
Schreiber et al. estimated the decrease in the window period attributable to the
increased sensitivity of a variety of tests, including those that detect the HIV
genome (29).
The use of window period estimates along with the incidence of new
infection is also applicable to other transfusion-transmitted agents, as exemplified
by the analysis published by the Retrovirus Epidemiology in Donors Study
(REDS) group (29). Lookback studies on other agents have not yet been per-
formed or if performed, have not been analyzed in a way that provides an estimate
of the window period.
Table 1 Published Risk of HIV Infection from Fully Screened and Tested Blood in
the United States
disparate. However, all of the estimates (prior to the implementation of HIV p24
antigen screening) fall within one order of magnitude. Further, there is a clear
trend over time towards lower risk estimates, commensurate with improve-
ments in test sensitivity and reductions in the frequency of test-positive dona-
tions. What is in fact remarkable is the similarity of the estimates, particularly
when those made during the same time interval are compared. In addition, recent
studies show, as might be expected, that there is considerable regional variation
in the residual risk in the United States, implying that national estimates will
vary from those developed for limited regions. One recent national estimate
of 1:420,000 (28) includes an adjustment for components infectious but sero-
negative for HIV, but that would nevertheless be withheld from transfusion
because of other test markers. This estimate is based upon a window period of 25
days, derived from lookback studies, subsequently corrected for the improved
sensitivity of current tests, and an incidence rate of 3.7 infections per 100,000
donations from repeat donors, representing 78% of donations. For first-time
donors, an estimate of 6.6 incident HIV infections per 100,000 was made.
Schreiber et al. (29) came up with a remarkably similar estimate of 1:493,000,
based upon careful studies in five large blood centers. Additionally, they esti-
mated the impact of additional testing, including that for HIV p24 antigen,
implying a current risk of 1:676,000 (29). As pointed out above, the risk of
disease and ultimately death from HIV infection is 100%. Consequently, every
potential infection may also be regarded as a potential death, albeit many years
after the original transfusion.
Overview of Infectious Disease 167
B. Hepatitis Viruses
One estimate of the risk of posttransfusion HBV infection (1:250,000 per unit) in
the United States is outlined above; it should be noted that this contrasts with a
reporting rate of one acute, clinical case per 30,000 recipients. In a preliminary
report from his study of cardiac surgery patients in Baltimore and Houston,
Nelson et al. suggest that the recipient seroconversion rate for anti-HBc implies
a per unit risk of about 1:2,500 after implementation of surrogate testing (ALT
and anti-HBc), with a further reduction to 1:25,000 after anti-HCV was im-
plemented (39). However, these findings have not been confirmed by additional
testing (particularly among the donors). Also, within the same study, there were
five seroconversions among 2,334 nontransfused patients. This rate did not differ
significantly from the overall seroconversion rate of 39 among the 9,449 trans-
fused patients. There are no contemporary prospective studies that define the
frequency of posttransfusion HBV infection. Surveillance studies now suggest
that only 1% of reported cases of hepatitis B have prior transfusion as a risk
factor (8).
Risk estimates based upon seroconversion rates and a window period of 59
days suggest that one unit in every 63,000 may be infectious for HBV (29). This
estimate is, however, dependent upon a window period that does not necessarily
reflect the true period of infectivity and upon adjustments that reflect the transient
nature of HBs antigenemia. Thus, this estimate should be regarded with some
cautionit seems likely that the risk is lower. Reasonable estimates of the
outcomes of HBV infection are that there will be clinically apparent acute disease
in 35% of cases, of which less than 0.5% will be fulminant, but of these, 60%
may result in death. Chronic disease appears to be very rare for adults, with a
long-term outcome in fewer than 5% of those infected.
The studies of Nelson et al. have been most instructive in the early
development of risk estimates for HCV infection. In their evaluation of transfused
cardiac surgery patients, they observed an incidence of one HCV infection per
3300 transfused components. This study represented donations tested by a first-
generation (c100-3) anti-HCV test and for anti-HBc and ALT elevations (12).
Subsequently, the study was extended and recipients (but not donations) were
tested using a so-called second-generation, multiantigen procedure. On this basis,
the residual risk was one infection per 1672 components (14) Kleinman and
others made an estimate of the potential improvement attributable to second-
generation testing. They based their estimate upon the increased frequency of
detection of HCV antibodies among donors and suggested that the residual risk
was between 1:2000 and 1:6000 (40). In Blajchman et als study, the frequency
of hepatitis C infection after the introduction of the first-generation test was 1.6
and 2.7 cases per thousand recipients of surrogate tested and nontested compo-
nents, respectively (19). A recent paper (41) points out that, as with HBV, care
168 Dodd
0.25% suffer fatal acute disease. Some 70% of infected individuals will have
chronic manifestations of infection. However, of all those infected, it appears that
up to 15% may eventually be diagnosed with liver failure and perhaps 3.5%
overall will die of liver disease after an incubation period of 20 years (38). There
does not seem to be any significant excess mortality during 1820 years of
follow-up of individuals originally diagnosed with posttransfusion NANB (37).
Follow-up studies do not provide reliable data beyond this period, although it
must be noted that in Japan some individuals have been shown to progress to liver
cancer (42).
There continues to be discussion about additional forms of viral hepatitis
that may be transmitted by transfusion. Although enteric viruses (such as HAV)
may be transmitted very infrequently as a result of collection of blood during an
asymptomatic preacute phase (43,44), there is also some evidence that post-
transfusion hepatitis (or at least, serum transaminase elevation) occurs in the
absence of any markers to known viruses (5,45,46). Most studies suggest that
these events are self-limited and that maximum ALT levels tend to be low.
Of great interest is Blajchman et al.s observation that this form of posttransfusion
hepatitis appears to occur among recipients of autologous transfusions as fre-
quently as it does among allogeneic blood recipients (19). Thus, great caution
should be used in attributing all (or perhaps any) such cases to an infectious agent.
An HCV-like virus, variously termed hepatitis G virus (47) or GB virus
type C (48), has been identified by molecular cloning and has been shown to be
associated with a minority (about 15%) of cases of residual posttransfusion
hepatitis. The agent is clearly transmissible by transfusion, but appears to have
little, if any, clinical relevance (4951).
Similarly, a DNA virus identified among three individuals with hepatitis
and termed TT virus (after one of the patients) has been shown to occur with
relatively high frequency and to be transmissible by transfusion. Again, however,
it has not proven possible to associate this virus with any evidence of liver
disease, and it appears to offer little, if any, risk to blood recipients (51a,51b).
C. Other Agents
The risk of HTLV infection was originally defined in Nelsons studies. In his
cohort, one infection was noted among the recipients of almost 70,000 tested
components (13). However, anti-HTLV-I tests are improving, particularly in their
ability to detect infection with HTLV-II, and as of 1997 two tests have been
licensed with a specific claim for detection of HTLV-II. The REDS group
estimated that window period infections would present a residual risk of about
1:641,000 (29). It is generally accepted that individuals who are seropositive for
HTLV-I have only about a 4% lifetime chance of developing either HTLV-
associated myelopathy or adult T-cell leukemia lymphoma (ATL). Although the
170 Dodd
former disease has been found in association with transfusion, this does not
appear to be the case for ATL. Published window period risk estimates for key
agents are outlined in Table 2.
Malaria is infrequently transmitted by transfusion in the United States: in a
1991 review, Nahlen et al. showed that the average reporting rate for clinically
apparent malaria is approximately one per 34 million units transfused (52);
surveillance data show that these rates have not changed appreciably through the
surveillance report of 1997 (53). The risk may be higher in countries with
populations having closer ties and more frequent travel to malarious areas.
An additional concern is the possibility of reestablishment of malaria in countries
from which it had previously been eliminated.
Infection with CMV is a relatively frequent outcome of transfusion, with
some studies indicating that 12% of blood units may be capable of transmitting
CMV. In almost all cases, however, such infection has little impact upon im-
munologically competent recipients but often has profound or fatal consequences
for immunocompromised patients. The risk of such outcomes is not well defined,
and increasing concentration on the provision of CMV seronegative or leuko-
reduced products has largely eliminated the concern for those patient populations
most at risk (20,54).
Another disease that has attracted some recent attention is Creutzfeldt-
Jakob disease (CJD). This is a prion disease with an extended incubation period.
It occurs with an incidence rate of about one case per million population annually,
and concern arises when a person with a history of blood donation is diagnosed
with this disease. The agent is clearly transmissible, although there is no current
evidence to show that it can be transmitted by transfusion. Indeed, case-control
NA = Not applicable.
aSee text.
and lookback studies have failed to show any evidence of such transmission
(5557). Animal studies suggest that the transmissible agent, when obtained from
brain tissue, may be infectious by parenteral inoculation. Conversely, intracranial
inoculation of blood from infected animals can also transmit disease. Recognition
that CJD had been transmitted by certain lots of human-derived growth hormone
has generated concern, and donors who received this material are now perma-
nently deferred (58), even though blood-to-blood transmission does not appear to
have been noted. This concern has been extended to recipients of dura mater and
to those with a family history of CJD. Despite the absence of any evidence of
transmission of CJD by transfusion, stringent requirements are now in place for
donor deferral and for recall of components of manufactured products collected
from donors subsequently found to be affected by, or judged to be at risk of, CJD.
These requirements are based solely upon the theoretical possibility of transmis-
sion. Additional concern has arisen, at least in the United Kingdom, as a result of
the apparent association between bovine spongiform encephalopathy (BSE) and
new variant CJD (nv CJD) in humans. Because the characteristics of the BSE
agent are not well known, specific measures have been proposed to reduce the
possible risk of transmission.
In the United Kingdom and some other countries, uniform leukoreduction
of components has been introduced, as it is thought that the agent of nvCJD is
more likely to be associated with leukocytes. In the United Kingdom, domes-
tically obtained plasma is no longer used for the manufacture of plasma deriva-
tives. This has led in the United States and Canada to consideration of procedures
to defer donors who have spent enough time in the British Isles to be considered
at some increased risk for nvCJD infection.
Finally, routine testing for markers of syphilis infection continues in most
countries. There appears to be essentially no risk of syphilis transmission by
transfusion at this time, and consequently no estimate is provided.
D. Bacterial Contamination
Since the adoption of closed plastic bag collection systems, bacterial contamina-
tion has not generally been regarded as a serious problem in transfusion medicine.
It is indeed true that bacterial contamination of red cell concentrates and fresh
frozen plasma is infrequent (5962). The major concern with red cells has been
sepsis or endotoxin shock resulting from transfusion of products with accumu-
lated high counts of Yersinia enterocolitica. In United States, such events occur
with a frequency of about one case per 3 million units transfused (63). A similar
rate of contamination with pseudomonads has been reported (64). These events
usually involve red cells that stored for 20 days or more. Bacterial outgrowth is
limited to organisms that can multiply at the 4C storage temperature of red cells.
It seems likely that the source of Y. enterocolitica is enteric infection in the donor,
172 Dodd
E. Laboratory Errors
As indicated above, laboratory errors are a possible source of risk to blood
recipients, inasmuch as improper performance of a test could lead to a false-
Overview of Infectious Disease 173
negative finding, but there is no clear measure of the frequency of such errors.
Systematic errors that might result in the failure of a complete test run are unlikely
to occur because of the need to obtain satisfactory results with test calibrators and
(where used) external controls, along with routine quality assurance measures.
Random errors such as sample transposition or omission may occur, although
modern, automated systems in place in blood centers should preclude such
problems. In order to affect a recipient, such an error would have to coincide with
the presence of an infectious sample. On the other hand, Young reported that 18,
27, and 55 HBsAg testing errors were reported to FDA in 1985, 1986, and 1988,
respectively (69). If every one of these reflected failure to identify an EIA-
positive product (which is by no means clear), then the error rate could have been
as high as 1855 per 18,000, assuming 18 million collections of whole blood and
plasma per year and an EIA-reactive rate of 0.1%. This would translate to an
overall error rate of 0.10.3%. In order to assess the risk of transmission of
disease, this error rate would have to be multiplied by the prevalence of true-
positive results. Thus, an error rate of 0.1% might lead to a risk of infection of
about one in 14 million for HIV (with a prevalence of six per hundred thousand);
the figure for HCV, with a prevalence of 0.16% would, however, be about one in
600,000. It is reasonable to assume that increased attention to transfusion safety
and quality assurance in testing over the past years will have resulted in low rates
of laboratory errors. Consequently, laboratory testing errors probably have no
significant contribution to the risk of infection from transfusion. Other widely
publicized errors certainly do occur but most often reflect failure to withhold a
safe, test-negative product because of a prior false-positive test result on the donor
of that unit (70).
individual who died within one year of the transfusion (26). This does not
necessarily mean that 50% of patients die, since those who did die may have
received more blood components. As discussed above, the overall mortality
among patients originally entered into posttransfusion hepatitis studies was
around 50% after 18 years of follow-up. However, a recent publication from
Vamvakas and Taswell suggests that posttransfusion mortality was not this high
in a community hospital system. However, posttransfusion mortality did vary
significantly according to age at transfusion (78). In summary, then, economic
estimates that define the overall costs of transfusion-associated disease should
recognize that up to 50% of the chronic disease outcomes will not contribute,
since the patients will have died before disease can be manifested.
nation remains unclear, but there have been no further reports of any outbreaks.
The B19 parvovirus is also nonenveloped and has been readily transmitted by
concentrates that had been subject to early, and less stringent, heat-treatment
regimens (81). It is unclear whether these transmissions led to any clinically
apparent disease. Currently, in some countries, fresh frozen plasma is also being
subjected to inactivation, either by methylene blue photoinactivation or solvent-
detergent treatment (8284). A possible disadvantage of the solvent-detergent
procedure is that it must be performed on a pooled product, consequently, risk
from enveloped viruses will be reduced or eliminated but at the cost of some
increase in the risk of exposure to HAV or B19. A solvent-detergenttreated
frozen plasma product has also been licensed and is in use in the United States.
Phase 4 (postlicensure) clinical studies showed that this product could indeed
transmit B19 parvovirus, as some lots were contaminated with a relatively high
titer of the virus. Preventive measures, including testing for viral DNA in final
products, have been implemented.
that can be gained, particularly for infections such as HCV where 40% or more
of the cases may be unaccompanied by identifiable risk (5,87).
Serological testing is unlikely to improve greatly, although it might be
possible to test for additional markers, as was the case for the HIV p24 antigen.
There is widespread anticipation that genome-based testing will offer significant
benefits. Indeed, the benefits of such testing have been projected (see Table 2).
It seems unlikely that any procedure for genome amplification testing of individ-
ual donations will be available in the immediate future but testing of pooled
samples has been implemented on a broad scale in Europe and the United States.
Although this application was implemented to reduce viral load in plasma for
further manufacture, it is clear that testing in small pools of 1624 will have
sufficient sensitivity to achieve significant improvement in the detection of HCV
window period donations and has the potential to detect at least some of the
remaining HIV window period donations. It will probably not have significant
effect upon HBV safety, however.
There may be some prospect for viral inactivation of cellular products, but
such procedures will almost certainly require the addition of chemicals or drugs
to the components; these, of course, will have their own risks, such as genotoxic-
ity. These risks may not outweigh the benefits to be gained. Finally, it will be very
important to consider emerging diseases in order to determine whether interven-
tions are necessary or appropriate. Ultimately, it must to be recognized that the
marginal gains that might be achieved by additional measures may not justify
the resources required to implement them.
REFERENCES
1. Dodd RY. The risk of transfusion-transmitted infection. N Engl J Med 1992;
327:419421.
2. McCullough J. The nations changing blood supply system. JAMA 1993; 269:2239
2245.
3. AuBuchon JP, Birkmeyer JD, Busch MP. Safety of the blood supply in the United
States: opportunities and controversies. Ann Intern Med 1997; 127:904909.
4. Alter MJ, Mares A, Hadler SC, Maynard JE. The effect of underreporting on the
apparent incidence and epidemiology of acute viral hepatitis. Am J Epidemiol 1987;
125:133139.
5. Alter MJ, Margolis HS, Krawczynski K, Judson FN, Mares A, Alexander WJ, Hu PY,
Miller JK, Gerber MA, Sampliner RE, Meeks EL, Beach MJ. The natural history of
community-acquired hepatitis C in the United States. N Engl J Med 1992; 327:1899
1905.
6. Alter MJ, Hadler SC, Margolis HS, Alexander WJ, Hu PY, Judson FN, Mares A,
Miller JK, Moyer LA. The changing epidemiology of hepatitis B in the United States.
Need for alternative vaccination strategies. JAMA 1990; 263:12181222.
178 Dodd
7. CDC. Hepatitis Surveillance Report No. 55. Atlanta: Centers for Disease Control and
Prevention, 1994: 136.
8. CDC. Hepatitis Surveillance Report No. 56. Atlanta: Centers for Disease Control and
Prevention, 1995: 133.
9. CDC. HIV/AIDS surveillance Report Vol. 10, No 2. Atlanta: Centers for Disease
Control and Prevention, 1999: 143.
10. Selik RM, Ward JW, Buehler JW. Trends in transfusion-associated acquired immune
deficiency syndrome in the United States, 1982 through 1991. Transfusion 1993;
33:890893.
11. Cohen ND, Munoz A, Reitz BA, Ness PK, Frazier OH, Yawn DH, Lee H, Blattner
W, Donahue JG, Nelson KE, Polk BF. Transmission of retroviruses by transfusion
of screened blood in patients undergoing cardiac surgery. N Engl J Med 1989;
320:11721176.
12. Donahue JG, Muoz A, Ness PM, Brown DE, Jr, Yawn DH, McAllister HA, Jr, Reitz
BA, Nelson KE. The declining risk of post-transfusion hepatitis C virus infection.
N Engl J Med 1992; 327:369373.
13. Nelson KE, Donahue JG, Muoz A, Cohen ND, Ness PM, Teague A, Stambolis VA,
Yawn DH, Callicott B, McAllister H, Reitz BA, Lee H, Farzadegan H, Hollingsworth
CG. Transmission of retroviruses from seronegative donors by transfusion during
cardiac surgery. A multicenter study of HIV1 and HTLV-I/II infections. Ann Intern
Med 1992; 117:554559.
14. Nelson KE, Donahue JG, Stambolis V. Post-transfusion hepatitis C virus infection.
Reply. N Engl J Med 1992; 327:16011602.
15. Aach RD, Szmuness W, Mosley JW, Hollinger FB, Kahn RA, Stevens CE, Edwards
VM, Werch J. Serum alanine aminotransferase of donors in relation to the risk of
non-A, non-B hepatitis in recipients. The Transfusion-Transmitted Viruses Study.
N Engl J Med 1981; 304:989994.
16. Koziol DE, Holland PV, Alling DW, Melpolder JC, Solomon RE, Purcell RH, Hudson
LM, Shoup FJ, Krakauer H, Alter HJ. Antibody to hepatitis B core antigen as a
paradoxical marker for non-A, non-B hepatitis agents in donated blood. Ann Intern
Med 1986; 104:488495.
17. Alter HJ, Purcell RH, Holland PV, Alling DW, Koziol DE. Donor transaminase and
recipient hepatitis. Impact on blood transfusion services. JAMA 1981; 246:630634.
18. Stevens CE, Aach RD, Hollinger FB, Mosley JW, Szmuness W, Kahn R, Werch J,
Edwards V. Hepatitis B virus antibody in blood donors and the occurrence of non-A,
non-B hepatitis in transfusion recipients. An analysis of the Transmission-Transmitted
Viruses Study. Ann Intern Med 1984; 101:733738.
19. Blajchman MA, Bull SB, Feinman SV. Post-transfusion hepatitis: Impact of non-A,
non-B hepatitis surrogate tests. Lancet 1995; 345:2125.
20. Bowden RA, Slichter SJ, Sayers M, Weisdorf D, Cays M, Schoch G, Banaji M, Haake
R, Welk K, Fisher L, McCullough J, Miller W. A comparison of filtered leukocyte-
reduced and cytomegalovirus (CMV) seronegative blood products for the preven-
tion of transfusion-associated CMV infection after marrow transplant. Blood 1995;
86:35983603.
21. Busch MP, Eble BE, Khayam-Bashi H, Heilbron D, Murphy EL, Kwok S, Sninsky
J, Perkins HA, Vyas GN. Evaluation of screened blood donations for human
Overview of Infectious Disease 179
Dodd RY, Petersen LR. Time course of detection of viral and serologic markers
preceding human immunodeficiency virus type 1 seroconversion: Implications for
screening of blood and tissue donors. Transfusion 1995; 35:9197.
35. Alter HJ. Youll wonder where the yellow went: a 15-year retrospective of post-
transfusion hepatitis. In: Moore SB, ed. Arlington, VA: AABB, 1987:5386.
36. Aledort LM. Consequences of chronic hepatitis C: a review article for the hematol-
ogist. Am J Hematol 1993; 44:2937.
37. Seeff LB, Buskell-Bales Z, Wright EC, Durako SJ, Alter HJ, Iber FL, Hollinger FB,
Gitnick G, Knodell RG, Perrillo RP, Stevens CE, Hollingsworth CG, NHLBI Study
Group. Long-term mortality after transfusion-associated non-A, non-B hepatitis.
N Engl J Med 1992; 327:19061911.
38. Koretz RL, Abbey H, Coleman E, Gitnick G. Non-A, non-B post-transfusion hepati-
tis: Looking back in the second decade. Ann Intern Med 1993; 119:110115.
39. Nelson KE, Ahmed F, Stambolis V, Ness PM, Yawn D, McAllister H. Incident
hepatitis C virus (HCV) and hepatitis B virus (HBV) infections in transfused
cardiac surgery patients: infection rates during different methods of donor screening.
Bethesda, MD: NIH Consensus Development Conference on Infectious Disease
Testing for Blood Transfusions. National Institutes of Health, 1995: 6769.
40. Kleinman S, Alter H, Busch M, Holland P, Tegtmeier G, Nelles M, Lee S, Page E,
Wilber J, Polito A. Increased detection of hepatitis C virus (HCV)-infected blood donors
by a multiple-antigen HCV enzyme immunoassay. Transfusion 1992; 32:805813.
41. Allander T, Gruber A, Naghavi M, Beyene A, Soderstrom T, Bjorkholm M, Grillner
L, Persson MAA. Frequent patient-to-patient transmission of hepatitis C virus in a
haematology ward. Lancet 1995; 345:603607.
42. Kiyosawa K, Sodeyama T, Tanaka E, Gibo Y, Yoshizawa K, Nakano Y, Furuta S,
Akahane Y, Nishioka K, Purcell RH, Alter HJ. Interrelationship of blood transfusion,
non-A, non-B hepatitis and hepatocellular carcinoma: analysis by detection of
antibody to hepatitis C virus. Hepatology 1990; 12:671675.
43. Hollinger FB, Khan NC, Oefinger PE, Yawn DH, Schmulen AC, Dreesman GR,
Melnick JL. Posttransfusion hepatitis type A. JAMA 1983; 250:23132317.
44. Noble RC, Kane MA, Reeves SA, Roeckel I. Posttransfusion hepatitis A in a neonatal
intensive care unit. JAMA 1984; 252:27112715.
45. Alter HJ, Purcell RH, Shih JW, Melpolder JC, Houghton M, Choo Q-L, Kuo G.
Detection of antibody to hepatitis C virus in prospectively followed transfusion
recipients with acute and chronic non-A, non-B hepatitis. N Engl J Med 1989;
321:14941500.
46. Alter HJ. New kit on the block: evaluation of second-generation assays for detection
of antibody to the hepatitis C virus. Hepatology 1992; 15:350353.
47. Linnen J, Wages J, Jr., Zhang-Keck ZY, Fry KE, Krawczynski KZ, Alter H, Koonin
E, Gallagher M, Alter M, Hadziyannis S, Karayiannis P, Fung K, Nakatsuji Y, Shih
JWK, Young L, Piatak M, Jr, Hoover C, Fernandez J, Chen S, Zou JC, Morris T,
Hyams KC, Ismay S, Lifson JD. Molecular cloning and disease association of
hepatitis G virus: a transfusion-transmissible agent. Science 1996; 271:505508.
48. Simons JN, Leary TP, Dawson GJ, Pilot-Matias TJ, Muerhoff AS, Schlauder GG,
Desai SM, Mushahwar IK. Isolation of novel virus-like sequences associated with
human hepatitis. Nature Med 1995; 1:564569.
Overview of Infectious Disease 181
A to patients with hemophilia by Factor VIII concentrates treated with organic solvent
and detergent to inactivate viruses. Ann Intern Med 1994; 120:17.
81. Azzi A, Ciappi S, Zakvrzewska K, Morfini M, Mariani G, Mannucci PM. Human
parvovirus B19 infection in hemophiliacs first infused with two high-purity, virally
attenuated Factor VIII concentrates. Am J Hematol 1992; 39:228230.
82. Lambrecht B, Mohr H, Knver-Hopf J, Schmitt H. Photoinactivation of viruses in
human fresh plasma by phenothiazine dyes in combination with visible light. Vox
Sang 1991; 60:207213.
83. Wieding JU, Hellstern P, Khler M. Inactivation of viruses in fresh-frozen plasma.
Ann Hematol 1993; 67:259266.
84. Horowitz B, Bonomo R, Prince AM, Chin SN, Brotman B, Shulman RW. Solvent/
detergent-treated plasma: a virus-inactivated substitute for fresh frozen plasma. Blood
1992; 79:826831.
85. Leape LL, Brennan TA, Laird N, Lawthers AG, Localio AR, Barnes BA, Hebert L,
Newhouse JP, Weiler PC, Hiatt H. The nature of adverse events in hospitalized
patients. Results of the Harvard Medical Practice Study II. N Engl J Med 1991;
324:377384.
86. Brennan TA, Leape LL, Laird NM, Hebert L, Localio AR, Lawthers AG, Newhouse
JP, Weiler PC, Hiatt HH. Incidence of adverse events and negligence in hospitalized
patients. Results of the Harvard Medical Practice Study I [see comments]. N Engl J
Med 1991; 324:370376.
87. Alter MJ, Hadler SC, Judson FN, Mares A, Alexander WJ, Hu PY, Miller JK, Moyer
LA, Fields HA, Bradley DW, Margolis HS. Risk factors for acute non-A, non-B
hepatitis in the United States and association with hepatitis C virus infection. JAMA
1990; 264:22312235.
9
Blood Donor Screening and
Supplemental Testing
Principles, Procedures, and Consequences
Jay E. Valinsky
New York Blood Center, New York, New York
I. INTRODUCTION
Blood transfusion has become increasingly safe worldwide since the introduction
of screening tests for transfusion-transmitted infectious diseases (TTD) (1) and
an extensive series of other measures to minimize the risk to recipients of blood,
blood components, and derivatives. Two events triggered dramatic improvements
in blood safety in the United States in the 1970s: the conversion to all-volunteer,
unpaid donors of blood for transfusion (2) and the widespread introduction of
tests for hepatitis B surface antigen that could be applied at the level of mass
screening (3). Cases of posttransfusion hepatitis dropped from about 25% of
recipients to about 5% of recipients.
The repertoire of blood screening tests for TTD expanded following the
introduction of tests for HIV-1 in the mid-1980s, screening tests for HIV, HTLV,
HCV, and HIV-1 p24 antigen, and tests of surrogate markers for liver dam-
age/disease (ALT and anti-HBc). Incremental improvements in blood safety
accompanied the initial implementation of each of these tests, and further
improvements were seen with increasingly more sensitive generations of tests.
Nucleic acid amplification testing (NAT) for HCV and HIV, and potentially other
transfusion-transmitted infectious agents, promises to reduce the already low
prevalence of these markers in the donor population and, by extension, the
exposure of transfusion recipients even further.
It should be emphasized that the ongoing improvements in blood safety did
not result solely from the introduction of new screening tests, but also by (a) more
carefully defining donor eligibility; (b) directly questioning donors about risk
185
186 Valinsky
behavior; (c) the establishment of registries of deferred donors; (d) the quarantine
of products until analysis of test results was complete and process controls
permitted labeling and release to inventory; (e) the introduction and enforcement
of current good manufacturing practices (cGMP); and (f) extensive monitoring
and investigation of adverse incidents, errors, and accidents (4).
All blood and plasma collected in the United States is screened for a variety
of infectious disease markers as well as ABO group and Rh type (up to 14
screening tests). A list of current screening tests is provided in Table 1. The U.S.
Food and Drug Administration (FDA) requires many of these screening tests for
the qualification of products and donors. Others, like assays for alanine amino-
transferase (ALT) and anti-hepatitis B core antibody, are still in use as surrogate
tests for nonA-E hepatitis or as product qualification tests for plasma destined
for further manufacture to blood derivatives. Still others (e.g., cytomegalovirus,
sickle cell trait) are used at the discretion of the blood centers as a means of
identifying products that may have specific clinical or therapeutic applications.
The focus of this chapter will be on the tests currently employed in the
United States for screening of volunteer blood doors for TTD and on the
disposition of donors and products as determined by test results. Other chapters
in this volume will directly address the issues of blood safety. General information
about the test algorithms, test kit qualification, controls, sensitivity and specificity,
and quality control will be followed by more specific information about the perfor-
mance and outcomes of each of the individual tests. The discussions in this chapter
are not intended to compare tests from different vendors nor to endorse the use of
specific tests. The tables that follow describe donor and product disposition reflect
requirements described in various FDA guidances and memoranda.
phases for performing EIA/ELISA, buffers, test reagents, and controls. Each kit
component is identified with a part number and has a defined expiration date. The
components are linked to a manufacturing master lot number with an independ-
ent expiration date. Components from different kit lots should not be inter-
changed, since test performance is based in part on the set of components linked
to a given master lot. Sample age, storage, and transport conditions are specified
in the manufacturers instruction circulars to ensure optimal test performance.
The EIA/ELISA tests used for detection of TTD markers use antigen or
antibody capture techniques (7). The antigen or antibody capture reagents (e.g.,
purified antibodies, monoclonal antibodies, purified antigens, cell or viral lysates,
or combinations thereof) are bound to a solid phase, typically a plastic support.
Different vendors have deployed a variety of test matrices, including 96-well
plastic microtiter/microwell plates, plastic beads, and microparticles, but the basic
principles are the same.
The EIA/ELISA tests are performed in sequential steps. Samples of plasma
or serum are incubated with matrix-bound capture reagents for a sufficient period,
at an appropriate temperature, to allow optimal binding of the analyte present in
the test sample to occur. Test controls (see below) are incubated simultaneously.
The matrix is then washed to remove unbound serum or plasma. The wash step
is followed by the addition of a conjugate reagent in one or several steps. These
reagents are typically enzyme-linked (e.g., horseradish peroxidase, alkaline phos-
phatase) anti-human Ig antibodies, enzyme-conjugated antigens, or biotin-avidin-
enzyme conjugates. During this incubation, the conjugate binds to the analyte
bound in the first step to the solid phase by the capture reagent. The matrix is
washed again to remove unbound conjugate. In the last step, or detection step,
reporter molecules or substrates that react with the bound enzyme conjugates are
added. Detection occurs as the specifically bound conjugates convert substrates
to chromogenic, fluorogenic, or chemiluminescent products or react with other
reporter molecules that can be detected spectrophotometrically. The signal (e.g.,
absorbance, relative fluorescence) produced in the test is proportional to the
amount of analyte initially present in the test sample. In qualitative tests, the
signal is evaluated relative to a threshold or cut-off value computed from
control values (see below).
The use of external controls has been somewhat controversial (8) because:
1. The performance characteristics of some of the external control
reagents in current use are not well defined. There is presently no
requirement for FDA clearance of these reagents, nor is there an abso-
lute requirement that they be manufactured under cGMP conditions.
2. External control values are sometimes used to define test performance
characteristics that exceed the manufacturers specifications. For exam-
ple, quantitative criteria for external control performance (e.g., controls
must perform within certain ranges of S/CO to be valid) have been
applied to semi-quantitative or qualitative tests.
3. There remain questions about how frequently external controls should
be run during the day (e.g., with each set of manufacturers controls,
with a batch of specimens, twice daily).
The Health Care Financing Administration (HCFA) has recently published rules
for the use of positive and negative external controls for in vitro diagnostic tests
(9). In commenting on these rules, FDA notes that the Clinical Laboratory
Improvement Amendments rules for use of these controls should be followed
but cautions that the use of external controls goes beyond the claims of the
manufacturers package inserts, that the manufacturers guidelines must be fol-
lowed, and that external controls must not be used as substitutes for the calibrators
included in the test kits (10).
mens must be retained and the samples subjected to the rest of the test algorithm.
The nonreactive (negative) samples should be retested (10). The procedure used
to assess the validity of the external control should be designed cautiously, so as
not to exceed the specifications of the test set by the manufacturer. Several
attempts have been made to establish performance rules for external controls
[e.g., Westgard rules (11) or other standard deviation rules]. However, these
statistical process control methods are not strictly applicable to the semi-
quantitative or qualitative tests used in donor screening. If the limits are too
stringent (e.g., values for the external control must be within 2 standard deviations
of mean), the frequency of failed runs due to random error alone is unreasonably
high (nearly 5%). If the limits are too relaxed (e.g., 3 standard deviations from
the mean), it is likely that a considerable percentage of run failures may go
undetected. Thus, it is more appropriate to apply a pass/fail system to qualify
external control performance for a particular run. Nonetheless, statistical process
control methods (e.g., control charts) should be applied rigorously to track shifts
or trends in laboratory performance using both the external and manufacturers
control values.
Lot acceptance criteria should also be established in each laboratory.
Upon receipt, a new master lot of test reagents should be quarantined pending
qualification for use. Lot acceptance may involve testing the new lot of reagents
against a panel of specimens that challenges the performance of the new lot in
the specific laboratory environment. The current lot should be tested in parallel.
This is an important step, since considerable variation in sensitivity is often
observed among master lots, even though they all meet manufacturing and
FDA specifications. The external controls should be included in the lot accep-
tance panel. This is most important for the positive external control, since it is
weakly reactive and may give false-negative results if the sensitivity of the new
lot is lower than the current lot. For purposes of tracking and trending the
performance of the external controls, it is also recommended that they be
requalified with each new master lot of reagents and that statistical parameters
(e.g., mean, standard deviation) be recalculated. This may be done conveniently,
for example by testing 20 replicates of the controls as part of lot validation.
As part of a quality improvement plan and to complement these quality control
measures, laboratories should actively participate in certified proficiency testing
programs managed by external organizations (e.g., College of American Pathol-
ogists, state health departments).
example, one test may be considered more sensitive if it is able to detect the
presence of an analyte in a sample when the other does not. On the other
hand, one test may be considered more sensitive if it can detect an analyte at
higher dilution. While the less sensitive of the two tests may detect the pres-
ence of low levels of analyte, it may still not detect all infected individ-
uals, thereby leading to false-negative results. Thus, while both definitions
apply, sensitivity, in the context of donor screening, is the ability of the assay
to identify true positives (TP) (e.g., infected individuals) in the test population,
while minimizing the number of false negatives (FN). The sensitivity of most
commercially available test kits currently exceeds 98%. Because the emphasis
is on the detection of as many positive donors as possible, most existing EIA
tests for donor screening produce a significant number of false-positive results
(see below).
As test sensitivity increases, the ability to identify individuals at earlier
stages of infection also improves. This reduces the time between exposure/infection
and detection [i.e., the window period (12)]. It is important to note that the length
of the window period is more a function of the sensitivity of the test than it is of
the disease. This has been shown clearly in the cases of HIV (13) and HCV
(14,15), in which the window period has decreased markedly with improvements
in test sensitivity in progressive generations of test kits.
The counterpoint of test sensitivity is specificity. Specificity can be defined
as the ability of a test to identify noninfected individuals in a population (true
negatives, TN), thereby avoiding false positives (FP). The specificity of most
commercially available EIA test kits exceeds 99%. It should be remembered that
sensitivity and specificity are essentially reciprocal values.
A convenient means of comparing tests, which takes both sensitivity and
specificity into account, is test efficiency. This is the ability of an assay to
correctly identify positives and negatives in the test population.
The predictive value (positive or negative) of a test is a semi-quantitative
assessment of the value of the test that takes the actual population prevalence into
account. Thus, for a given specificity and sensitivity, the value of tests may differ
depending on the populations tested, e.g., low-risk blood donors versus high-risk
populations in a sexually transmitted disease clinic. In the case of blood donor
testing, the negative predictive value of a test may be the more valuable
parameter, since it estimates the ability of the test to detect false negatives.
Equations for the calculation of the parameters defined in this section are
found in Table 2.
The ability of these serological assays for infectious disease markers
to detect antibodies or antigens may vary regionally, reflecting the genotypic
distribution of the infectious agent (16,17). In some instances it has been
Blood Donor Screening and Testing 193
Parameter Calculation
shown that mutations in the infectious agent may go undetected in some tests
(18,19).
Screening result
Result Suitable for Interdict Sample to Product Product
Analyte Initial Repeat interpretation transfusion shipment confirmation label disposition
Screening test result Confirmatory result Surveillance Deferral type Reentry available
196
The combination of a HIV-1/2 RR and a HIV-1 p24 antigen RR on any single or combination of donations will prevent reentry.
The combination of a HBsAg RR and HBc RR on any single or combination of donations will result in permanent deferral.
Table 6 Hospital In-Date Product Retrieval and Lookback
1. Western Blots
The Western blot is one of the most widely used tests for the confirmation of the
presence of antibodies to retroviruses. A Western blot assay is typically prepared
from partially purified viral proteins derived from viral lysates, recombinant
proteins, peptides, or combinations thereof. The viral proteins are separated by
SDS-polyacrylamide gel electrophoresis according to apparent molecular weight.
Following electrophoretic separation, the proteins are arrayed in the gel as
concentrated bands. The proteins are then transferred electrophoretically from
the polyacrylamide to strips of nitrocellulose paper, where the proteins/peptides
are stabilized and stored until use. The nitrocellulose strips, therefore, contain an
image of the proteins as originally displayed in the polyacrylamide gel.
To perform the test, samples of serum or plasma are incubated with the
nitrocellulose test strips and processed in a manner similar to an EIA. Antibodies
in the test sample bind to specific proteins or antigens arrayed on the nitro-
cellulose strip. Binding is revealed through a series of reactions with enzyme-
antibody conjugates and substrates. Colored products produced by the enzymatic
reaction precipitate and bind to the nitrocellulose strips. The identity of the
antibody binding sites is established both by the position of the stained protein
band on the strip (i.e., apparent molecular weight) and by the reactivity estimated
by the staining intensity. The specificity of the test arises from this specific
binding of antibody to recognizable virus-associated antigens, and the sensitivity
of the test arises, in part, from the concentration of the viral antigens in
electrophoretic bands. The test kits include positive and negative control samples
that give predictable patterns of antibody binding and intensity that can be used
to calibrate the position and reactivity of the test samples.
Typically results are reported as positive (POS) if there is a pattern of
antibody binding to diagnostic bands specific for the virus in question or negative
(NEG) if there are no specific binding patterns or no antibody binding at all.
Results may also be indeterminate (IND) if there are binding patterns that do not
meet the criteria for positive. Failure of the control reagents to produce the
expected pattern of binding and/or intensity invalidates the entire run. A discussion
of Western blot tests for particular viral markers can be found in subsequent sections.
2. Immunofluorescence Assays
An alternative to the Western blot for the detection of viral antibodies is the
Immunofluorescence assay (IFA). Unlike the Western blot, IFA does not identify
specific antibodies. A positive result simply indicates whether antibodies to the
virus in question were present in the test sample. Immunofluorescence assays
have been used as confirmatory tests for HIV, HTLV, and syphilis.
In this test, which is basically an indirect immunofluorescence assay, cells
infected with a particular virus are deposited and fixed in wells etched or
Blood Donor Screening and Testing 199
circumscribed on a glass microscope slide. The fixed cells are then incubated with
test serum or plasma. After a series of washes to remove unbound test sample,
the cells are incubated with a fluorescently tagged anti-human IgG. Viral antigens
present intracellularly are revealed by fluorescence microscopy. The specificity
of the test is assessed by comparing the immunofluorescence intensity and
patterns in the infected cells with those observed in noninfected cells treated with
the same test sample. Staining intensity and patterns produced by the test samples
can also be compared with those produced by the positive and negative controls.
The fluorescence intensity is usually scored on an arbitrary scale of 0 to 4+.
Additional quality controls include evaluation of patterns of fluorescence (e.g.,
membrane vs. ctyoplasmic, hazy vs. well-demarcated fluorescence).
incubations. If the antigen is present in the neutralized sample, some or all will
bind to the antibody in solution and form an antigen-antibody complex. Antigen
in the complex will be unavailable for binding to the antibody on the solid phase.
The signal in the EIA reaction will be reduced relative to the sample incubated
with the control antibody. The percentage of inhibition or neutralization can then
be determined by comparing the neutralized and nonneutralized values. The
sample is considered confirmed positive if assay-specific threshold values are
achieved (see below). It has recently been shown that neutralization assays may
be prone to artifacts that produce false-positive (23,24) and/or indeterminate
results (25).
Gene product
gp41
gp120 p24 p31 (e.g. p70)
gp160
None None None None Negative
Any p24 Any Any or none
Any p24 Any Any or none Positive
Any 2 None Any or none Any or none
Any Other Pattern Any or none Indeterminate
None None None Any Indeterminate
aAdditional reactivities for gag (e.g., p17, p26, and p55) and pol (e.g., p56 and p66) gene products
may also be present in band patterns associated with a fully reactive specimen. However, these are
not diagnostic bands defined in the CDC criteria nor in the manufacturers package inserts. Nonviral
bands (NVB) commonly appear in the p70 region of the Western blot strip but may also appear in
other regions. NVBs do not appear to be associated with HIV-specific antigens. Nonetheless, they are
recorded during the reading of the strips. Their appearance can affect the interpretation of the strip in
only one case, namely that instance in which NVBs appear on a strip devoid of HIV diagnostic bands.
Under current guidelines, this strip must be interpreted as Indeterminate.
204 Valinsky
Samples that are reactive in the HIV-2 EIA may be subjected to an HIV-2
Western blot. Currently these blots are available for research use only. Valid test
results for this assay are:
The algorithm for final interpretation of the HIV-1/2 testing, based on the
combined results of the Western blot or IFA and EIA tests, is presented in Table 8.
HIV-1/2 HIV-1 WB
EIA or IFA HIV-2 EIA HIV-2 WB Final interpretation
Recent data suggest that many donors described as positive for HTLV
using these algorithms may be false positive (47,48). Because of this, and
because of the unavailability of licensed confirmatory tests, an algorithm was
proposed in which a second FDA-licensed EIA test is used to confirm the
presence of HTLV antibodies in a given specimen (49). This test algorithm is
based on results that suggest that many specimens repeatedly reactive in one EIA
may not be reactive in a second licensed test. Thus, only those specimens that
are reactive in both tests are considered positive for HTLV. Although it is
208 Valinsky
not an obligatory part of the algorithm, additional testing using the investiga-
tional or research use only Western blot is suggested for those samples positive
in the two EIAs. This algorithm does not allow discrimination of HTLV-I and
HTLV-II. Additional testing using research use only PCR tests may be valuable
in this regard.
G. Alanine Aminotransferase
Alanine aminotransferase (ALT) an enzyme involved in amino acid metabolism,
is found in highest concentrations in the liver and kidney. The enzyme catalyzes
the conversion of alanine and -aminoglutaric acid to pyruvic acid and glu-
tamic acid.
The appearance of the enzyme in the serum is often taken as an indicator
of tissue damage. ALT testing of blood donors was initiated as a surrogate marker
for non-A, non-B hepatitis in the mid- to late 1980s. Its value has been reduced
significantly following the introduction of specific tests for HCV (55). Neither
AABB nor FDA requires donor screening for ALT. However, ALT screening is a
requirement for the shipment of recovered plasma for further manufacture in the
countries of the European Union (56).
A variety of tests have been used to measure the ALT activity in serum.
These generally take advantage of coupled enzyme reactions utilizing the prod-
ucts of the ALT reaction in subsequent enzymatic reactions, which, in turn, lead
to the formation of chromogenic products that can be detected spectrophotomet-
rically. Both kinetic and endpoint tests have been used. The cut-off for acceptable
ALT activity is defined in terms of either studies on the distribution of ALT
activities in the test population or by using the normal values defined by the
manufacturers package insert (57). The valid results for the ALT test are:
Blood Donor Screening and Testing 211
H. Syphilis Testing
Serological tests for syphilis (STS) were the first infectious disease marker tests
applied to blood for transfusion. Syphilis testing was essentially the first test for
transfusion-transmitted diseases. Currently the STS employed for screening blood
donors include manual reagin tests [e.g., rapid plasma reagin tests (RPR)] or
microhemagglutination (MHA) tests antibodies to Treponema pallidum. These
tests appear in large measure to identify biological false positivesindividuals
who retain antibodies for many years following exposure and a much smaller
subset of individuals who may have active or untreated syphilis (58). Transmis-
sion of syphilis by blood transfusion is an extremely rare occurrence. STS is still
required for donor screening.
The RPR test detects the presence of reagin in the serum or plasma of
infected individuals using a cardiolipin antigen bound to carbon particles.
When the reagin is present, an agglutination reaction occurs, which can be scored
visually. Test results are either reactive or nonreactive. Reactive results should be
interpreted cautiously, should be confirmed in a quantitative test and should be
reviewed in the context of health history. Confirmation using a quantitative test
is typically not part of the screening algorithm, but rather a treponemal test,
FTA-ABS (see below), is used.
Alternative tests, either EIA or MHA-TP tests, have been automated for
mass screening and are more appropriately deployed in the context of donor
screening than the reagin tests. The MHA-TP type tests, which are in wide use,
analyze serum or plasma samples for the presence of antibodies to T. pallidum.
The presence of antibody is detected in an agglutination reaction measured by
light scattering or other photometric methods. The results are typically reported
as reactive or nonreactive (59). In some cases, results may be reported as
indeterminate. These are technically treated as reactive, and samples are reflexed
to the confirmatory algorithm.
Reactive findings in the screening tests are confirmed using the fluores-
cent treponemal antibody absorption test (FTA-ABS). In this assay, test serum
is treated with a sorbent derived from a Reiter strain culture of T. pallidum
to absorb nonspecific antibodies. This absorbed sample is then incubated
with T. pallidum fixed to microscope slides. If the sample contains antibody,
it will bind to the fixed cells. Specific antibody binding is detected by fluo-
rescence microscopy following incubation with FITC-conjugated anti-human
212 Valinsky
antibody. Test results are compare to positive and negative control sera. Valid
results are reactive or nonreactive based on staining intensity. Because the
false-positive rate for blood donor samples is significant (6062), additional
testing, using RPR or other assays, may be employed for counseling pur-
poses. The composite of interpretations of syphilis results can be found in
Table 9.
FTA-ABS
Screening test result result RPR result Interpretation
Table 10 Nucleic Acid Amplification Testing for HCV and HIV RNA
1. Possible Results
Suitable for
NAT HCV or HIV Result Interpretation transfusion
PCR Assaya
NEG Negative Yes
POS Positive No
Pos Pool/Indiv. Negb Inconclusive No
Pos Pool/QNSc Inconclusive No
TMA Assayd
NEG Negative Yes
POS Positive No
HIV POS Positive for HIV-1 RNA No
HCV POS Positive for HCV RNA No
NAT POS Positiveno discrimination No
between HIV and HCV
aSpecimens are tested in primary pools of 24 plasma samples. If the primary pool is
negative, all 24 samples are reported negative. If the primary pool is positive, specimens
are retested in 4 pools of 6 samples each. If a positive pool(s) is (are) identified, specimens
are retested as individual samples. The result reported is the final result. Hold implies that
test results during the pool resolution were inconclusive. QNS implies that the pool
resolution could not be completed.
bThis result reflects a positive test on the primary pool, but negative results in testing of
negative, all 16 samples are reported negative. If the primary pool is positive, all 16
specimens are retested individually. If one or more specimens are positive, they are
subjected to discriminatory TMA assays to distinguish between HIV and HCV. All negative
samples are released. The result reported is the final result following discriminatory testing.
In those cases where the discriminatory assay fails, the result is reported as positive.
Product disposition
Result Interdict
interpretation shipment Product label RBC/PLT Plasma
Donor Recipient
NAT result for Deferral type notification lookback
HCV
Negative None None No
Positive 1 yr unless seroconverts to In person Yes
HCV antibody POS
Pos pool/Indiv neg Surveillance In person No
Pos pool/QNS 1 yr or until retest negative In person No
HIV
Negative None None No
Positive 6 mo unless seroconverts In person Yes
to HIV antibody POS
Pos pool/Indiv. neg Surveillance In person No
Pos. pool/QNS 6 mo or until retest negative In person No
Note: In those cases where the TMA discriminatory assay is inconclusive, the donors are managed as
if the result were positive.
REFERENCES
1. Busch M, Chamberland M, Epstein J, et al. Vox Sang 1999; 77:6776.
2. Eastlund T. Monetary blood donation incentives and the risk of transfusion transmit-
ted diseases. Transfusion 1998; 38:874882.
3. Alter HJ, Holland PV, Purcell RH, et al. Post-transfusion hepatitis after exclusion of
commercial hepatitis B antigen positive donors. Ann Int Med 1972; 77:691699.
4. U.S. Food and Drug Administration. Guidance for Industry. Quality assurance in
blood establishments. Rockville, MD: CBER Office of Communication, Training and
Manufacturers Assistance, 1995.
5. Giles RE, Perry KR, Parry JV. Simple rapid test devices for anti-HIV screening: Do
they come up to the mark? J Med Virol 1999; 59:104109.
6. Mvere D, Constantine M, et al. Rapid and simple hepatitis assays: encouraging results
from a blood donor population in Zimbabwe. Bull WHO 1996; 74:1924.
7. Constantine NT, Callahan JD, Watts DM. Retroviral Testing: Essentials for Quality
Control and Laboratory Diagnosis. Boca Raton, FL: CRC Press, 1992.
8a. Linden JV, Wethers J, Dressler KP. Controversies in Y medicine: use of external
controls in transmissible disease testing: pro. Transfusion 1994; 34:550551.
8b. Epstein JS. Controversies in transfusion medicine: use of external controls in
transmissible disease testing: con. Transfusion 1994; 34:552553.
9. Code of Federal Regulations, 42CFR Part 493. Washington, DC: U.S. Government
Printing Office, 2000.
10. U.S. Food and Drug Administration. Draft Guidance for Industry: revised recommen-
dations for the invalidation of test results when using licensed and 510(k) cleared
216 Valinsky
blood borne pathogen assays to test donors. Rockville, MD: CBER Office of
Communication, Training and Manufacturers Assistance, 1999.
11. Green GA 4th, Carey RN, Westgard JO, et al. Quality control for qualitative assays:
quantitative QC procedure designed to assure analytical quality required for an
ELISA of hepatitis B surface antigen. Clin Chem 1997; 43:16181621.
12. Schreiber GB, Busch MP, Kleinman SH, Korelitz JJ. The risk of transfusion-
transmitted viral infections. The Retrovirus Epidemiology Donor Study. N Engl J
Med 1996; 334:16851690.
13. Busch MP, Lee LL, Satten GA, et al. Time course of detection of viral and serologic
markers preceding human immunodeficiency virus type 1 seroconversion: impli-
cations for screening of blood and tissue donors, Transfusion 1995; 35:9197.
14. Kleinman S, Busch MP, Korelitz JJ, Schreiber GB. The incidence/window period
model and its use to assess the risk of transfusion-transmitted human immunodefi-
ciency virus and hepatitis C virus infection. Transfus Med Rev 1997; 11:155172.
15. Vrielink H, Reesink HW, van den Burg PJ, et al. Performance of three generations of
anti-hepatitis C virus enzyme-linked immunosorbent assays in donors and patients.
Transfusion 1997; 37:845849.
16. Lee JH, Roth WK, Zeuzem S. Evaluation and comparison of different hepatitis C
virus genotyping and serotyping assays. J Hepatol 1997; 26:10011009.
17. Cheingsong-Popov R, Osmanov S, Pau CP, et al. Serotyping of HIV type 1 infections:
definition, relationship to viral genetic subtypes, and assay evaluation. UNAIDS
Network for HIV-1 Isolation and Characterization. AIDS Res Hum Retroviruses
1998; 14:311318.
18. Schable C, Zekeng L, Pau CP, et al. Sensitivity of United States HIV antibody tests
for detection of HIV-1 group O infections. Lancet 1994; 344:13331334.
19. Jongerius JM, Webster M, Cuypers HT, et al. New hepatitis B virus mutant form in
a blood donor that is undetectable in several hepatitis B surface antigen screening
assays. Transfusion 1998, 38:5659.
20. Zaaijer HL, van Rixel T, van Exel-Oehlers P, et al. New anti-human immunodefici-
ency virus immunoblot assays resolve nonspecific western blot results. Transfusion
1997; 37:193198.
21. Kleinman SH, Busch MP, Stramer SL, Watanabe K. Use of HIV-1/HIV-2 recombinant
immunoblot assay (RIBA) to resolve infection status of donors with possible false
positive HIV Western blot (WB) (abstract). Transfusion 1997; 37(suppl):47S.
22. Vrielink H, Zaajier HL, van der Poel CL, et al. New strip immunoblot confirmation
of HTLV-I/II infection. Vox Sang 1996; 70:114116.
23. Kleinman S, Busch M, Rawal B, Glynn S. Evaluation of HBsAg neutralization test
positive (Neut POS) donors with negative (NEG) anti-HBc (abstract). Transfusion
1998; 38(suppl):92S.
24. Stramer SL, Aberle-Grasse J, Bordsky JP, et al. US blood donor screening with p24
antigen (Ag): one year experience (abstr). Transfusion 1997; 37 (suppl):1S.
25. Strauss D, Valinsky JE, Kessler D, et al. Specificity of HIV-1 p24 antigen neutraliza-
tion (abstract). Transfusion 1996; 36(suppl):38S.
26. Murthy KK, Henrard DR, Eichberg JW, et al. Redefining the HIV-infectious window
period in the chimpanzee model: evidence to suggest that viral nucleic acid testing
can prevent blood-borne transmission. Transfusion 1999; 39:688693.
Blood Donor Screening and Testing 217
27. Busch MP, Kleinman SH, Jackson B, et al. Committee report. Nucleic acid amplifi-
cation testing of blood donors for transfusion-transmitted infectious diseases: report
of the Interorganizational Task Force on Nucleic Acid Amplification Testing of Blood
Donors. Transfusion 2000; 40:143145.
28. U.S. Food and Drug Administration. Guidance for Industry: in the manufacture and
clinical evaluation of in vitro tests to detect nucleic acid sequences of human
immunodeficiency viruses types 1 and 2. Rockville, MD: CBER Office of Commu-
nication, Training and Manufacturers Assistance, 1999.
29. Tilston P, Morris DJ, Klapper PE, Corbitt G. Commercial assay for hepatitis C virus
RNA. Lancet 1994; 344:201202.
30. MacDonough SH, Giachetti C, Yang Y, et al. High throughput assay for the simulta-
neous or separate detection of human immunodeficiency virus-1 (HIV-1) and hepa-
titis type C virus (HCV). Infusionther Transfusionmed 1998; 25:1649.
31. Myers TW, Gelfand DH. Reverse transcription and DNA amplification by a Thermus
termophilus DNA polymerase. Biochemistry 1994; 30:76617666.
32. Code of Federal Regulations, Title 21 CFR Part 610. 610.45. Washington, DC: U.S.
Government Printing Office.
33. Menitove J, ed. Standards for Blood Banks and Transfusion Services. 19th ed.
Bethesda, MD: American Association of Blood Banks, 1999.
34. Food and Drug Administration. Memorandum: use of Fluorognost HIV-1 immuno-
fluorescent assay (IFA). Rockville, MD: CBER Office of Communication, Training
and Manufacturers Assistance, 1992.
35. Centers for Disease Control. Interpretive criteria used to report Western blot results
for HIV-1 antibody testing, MMWR 1991; 40:692695.
36. Kleinman S, et al. False-positive HIV-1 test results in a low risk screening setting
of voluntary blood donation. Retrovirus Epidemiol Donor Study, JAMA 1998;
280:10801085.
37. Sayre KR, Dodd RY, Tegtmeier G, et al. False-positive human immunodeficiency
virus type 1 western blot tests in non-infected blood donors. Transfusion 1996;
36:4552.
38. Human Immunodeficiency Virus Type 1 (HIV-1) Fluorognost HIV-1 IFA.
Manufacturers package insert. Vienna, Austria: Waldheim Pharmazeutika,
Ges.m.b.H, 1992.
39. Centers for Disease Control and Prevention. Update: HIV-2 infections among blood
and plasma donorsUnited States. JAMA 1995; 274:10071008.
40. U.S. Food and Drug Administration. Memorandum: recommendations for donor
screening with a licensed test for HIV-1 antigen. Rockville, MD: CBER Office of
Communication, Training, and Manufacturers Assistance, 1995.
41. Stramer S, Aberle-Grasse J, Bordsky J, et al. United States blood donor screening
with p24 antigen: (one year experience) (abstract). Transfusion 1997; 37(suppl):1S.
42. Food and Drug Administration. Guidance for industry: donor screening for anti-
bodies to HTLV-II. Rockville, MD: CBER Office of Communication, Training and
Manufacturers Assistance, 1997.
43. Vrielink J, Reesink HW, Zaaijer JL, et al. Sensitivity and specificity of four assays to
detect human T-lymphotropic virus type I or type I/II antibodies. Transfusion 1996;
36:344346.
218 Valinsky
44. Liu H, Sha M, Stramer S, et al. Sensitivity and specificity of human T-lymphotropic
virus (HTLV) types I and II polymerase chain reaction and several serologic assays
in screening a population with a high prevalence of HTLV-II. Transfusion 1999;
39:11851193.
45. Anderson DW, Epstein JS, Lee TH, et al. Serological confirmation of human
T-lymphotrophic virus Type I infections in healthy blood and plasma donors. Blood
1989; 74:25852591.
46. Gallo D, Penning LM, Hanson CV. Detection and differentiation of antibodies to
human T-cell lymphotropic virus types I and II by the immunofluorescence method.
J Clin Micro 1991; 29:23452347.
47. Busch MP, Kleinman SH, Stramer SL. False positive HTLV-I/II supplemental test
results in blood donors (abstr). Transfusion 1999; 39:105S.
48. Stramer SL, Layug L, Trenbeath J, et al. Use of a second EIA in an HTLV-1/HTLV-2
algorithm (abstr). Transfusion 1998; 38(suppl):81S.
49. American Association of Blood Banks. Association bulletin #99-9. Dual enzyme
immuno assay (EIA) approach for deferral and notification of anti-HTLV-I/II EIA
reactive donors. Bethesda, MD: American Association of Blood Banks, 1999.
50. Strauss D, Valinsky JE, Kessler D, Bianco C. Decreased specificity of HBsAg
confirmation as a result of decreased specificity of the screening test (abstract).
Transfusion 1994; 34(suppl):34S.
51. Busch MP, Tobler LH, Stramer S, et al. Yield of HCV EIA 3.0 vs EIA 2.0 in screening
of US blood donors (abstr). Transfusion 1997; 37(suppl):111S.
52. Skidmore S. Recombinant immunoblot assay for hepatitis C antibody. Lancet 1990;
335:1346.
53. Schifman RB, Rivers SL, Sampliner RE, Kramer JE. Significance of isolated hepatitis
B core antibody in blood donors. Arch Int Med 1993; 153:22612266.
54. Lok A. Hepatitis B infection: pathogenesis and management. J Hepatol 2000;
32(suppl):8997.
55. Cable R, Badon S, Pray C, Popovsky MA. Limited utility of alanine aminotransfer-
ase screening of hepatitis C antibody screened blood donors. Transfusion 1997;
37:206210.
56. Committee for Proprietary Medicinal Products. Plasma derived medicinal products:
position paper on ALT testing, CPMP/BWP/385/99, 1999.
57. American Association of Blood Banks. Association bulletin #95-7. FDA notification
requirement: ALT procedure change, change to the circular of information. Bethesda,
MD: American Association of Blood Banks, 1995.
58. Orton S, Liu H, Cable R, et al. Prevalence of circulating T. pallicum in STS+/F
TA-Abs+ blood donors (abstr). Transfusion 1999; 39(suppl):2S.
59. Larsen SA, Hunter EF, Kraus SJ. A manual of tests for syphilis. Washington, DC:
American Public Health Association, 1990.
60. Win N, Islam SI, Peterkin MA, Walker ID. Positive direct antiglobulin test due
to antiphospholipid antibodies in normal healthy blood donors. Vox Sang 1997;
72:182184.
61. Fosdick M, Winslow D, Larsen S. Clinical significance of MHATP as a confirmatory
test for the Olympus PK-TP system (abstr). Transfusion 1994; 34(suppl):49S.
Blood Donor Screening and Testing 219
62. Strauss D, Del Valle C, Valinsky JE, et al. Donor screening for syphilis using the
Olympus PK-TP assayimplications for donor notification and re-entry. Transfusion
1993; 33(suppl):38S.
63. Committee for Proprietary Medicinal Products. The introduction of nucleic acid
amplification technology (NAT) for detection of hepatitis C virus RNA in plasma
pools, addendum CPMP/BWP/390/97 to CP/BWP/269/95, 1998.
10
Bacterial Contamination
I. TRANSFUSION-TRANSMITTED BACTERIAL
INFECTION OF PLATELETS
Sepsis due to transfusion of bacterially contaminated platelets is the most com-
mon transfusion-transmitted disease, principally because of the fact that platelets
are stored for up to 5 days at 2024C (3). In the years 19861991 there was a
total of 182 transfusion-related deaths reported to the U.S. Food and Drug
Administration, (FDA) 29 of which were due to bacterially contaminated blood
products. Some 21 (72%) of these deaths were associated with the transfusion of
221
222 Spinelli and Brecher
bacterially contaminated platelets (2). Five fatalities were reported in both 1990
and 1991. Five to six million random platelet concentrates and single donor
apheresis are transfused per year in the United States (46). This would mean that
there is at least a one in a million chance of death due to sepsis per unit transfused.
However, it is widely suspected that platelet bacterial sepsis is frequently unrec-
ognized and thus underreported.
The contamination rate for platelets is approximately 1 in 1000 per individ-
ual unit (7,8). Single unit contamination is similar for both platelet concentrates
made from whole blood and single-donor apheresis concentrates, but the ultimate
risk of sepsis is likely to be 610 times greater with pooled random units, as there
is a 6- to 10-fold increased donor exposure (7,9). It has been estimated that as
many as 150 people per year in the United States suffer severe morbidity and or
mortality as a consequence of a platelet transfusion (2).
In a recent study of symptomatic bacteremia following platelet transfusion
in 161 bone marrow transplant recipients in Hong Kong, it was found that 1 in
2000 units of platelet concentrates were bacterially contaminated. This translated
to 1 in 350 pooled platelets being contaminated. Of those patients who were
febrile (elevation of temperature of 1C) following platelet transfusion, 1 in 4
(27%) were found to have received a bacterially contaminated unit. Of those
found to have a 2C rise in temperature following a platelet transfusion, 50%
were found to have received a bacterially contaminated unit (10). In this multiply
transfused patient population, the chance of receiving a bacterially contaminated
platelet was 1 in 16. Of the 10 patients who are known to have received a
bacterially contaminated unit, 4 suffered from septic shock.
It is felt that the incidence of sepsis due to red cell transfusions has
decreased over the last 30 years (11), most likely due to the introduction of
disposable collection sets and bags in the 1960s. Yet, the frequency of bacterial
sepsis due to platelet transfusion has increased, largely due to increasing use of
this component and prolonged storage time at 20-24C.
B. Clinical Presentation
The clinical sequelae of the transfusion of bacterially contaminated platelets
may range from asymptomatic to mild fever (which may be indistinguishable
from a nonhemolytic transfusion reaction) to acute sepsis, hypotension, and death.
The presentation is much more varied and often less severe than that of pa-
224 Spinelli and Brecher
1986 and 1991 were older than 25 days (2). However, cases of Yersinia red cell
sepsis have been reported following as little as 714 days of storage (29,33).
Serratia marcescens has been implicated in the contamination of plastic
blood containers manufactured in a plant in Belgium, which affected several
Danish and Swedish transfusion centers. The outbreak was thought to involve
the manufacturing process and resulted in the closing of a blood bag manufac-
turing plant (34,35). Serratia liquefaciens has been recently recognized as both a
red cell and a platelet contaminant in the United States (L. Bland, personal
communication). Other gram-negative organisms that have been repeatedly asso-
ciated with red cell contamination are Pseudomonas and Enterobacter spp.
(12,24,36,37).
C. Clinical Presentation
Symptoms associated with transfusion of contaminated red cells are more severe
and rapid in onset than those caused by an infected platelet transfusion. Patients
frequently develop high fever (temperatures as high as 109F have been observed)
and chills during or immediately following transfusion (42,43). From 1987 to
February 1996, 20 recipients of Yersinia-infected red cells in 14 states were
reported to the CDC. Twelve of the 20 recipients died in 37 days or less following
transfusion. The median time to death was 25 hours! Of the 7 who developed
disseminated intravascular coagulation (DIC), 6 died. Signs, symptoms, and
complications are summarized in Table 2 (44).
In an anesthetized patient, hypotension, oozing, oliguria/anuria, and fever
should alert the anesthesiologist to the problem. Affected recipients may also
experience nausea, vomiting, and diarrhea. In severe instances, DIC, vascular
collapse, and death can rapidly ensue. If a septic transfusion reaction is suspected,
the transfusion should be discontinued immediately and not restarted. The mini-
mum number of organisms necessary to cause clinical symptoms is not known,
but concentrations of 108 CFU/mL or greater have been associated with severe
reactions and death (11).
226 Spinelli and Brecher
Chills 16 80
Fever 14 70
Hypotension 13 65
DIC 7 35
Death 12 60
Source: Ref. 60.
A. Detection
There is no universally accepted test, method, or device used in the detection of
bacterially contaminated blood product. Ideally, the test must be sensitive (pref-
erably to 104105 CFU/ml), specific, inexpensive, simple to use, capable of
Bacterial Contamination 227
detecting the commonly implicated organisms, adaptable for new bacterial threats,
and usable in a hospital environment. Approaches currently under investigation
are summarized below.
1. Bacterial Staining
Stain of a smear immediately prior to transfusion is sensitive to approximately
106 CFU/mL for gram-positive and 108 CFU/mL for gram-negative organisms
(51,52). Gram stain is most often employed, although Wrights stain and acridine
orange have also been recommended (53,54). This test lacks the necessary
sensitivity to detect all clinically significant bacterial contamination and has been
associated with both high false-positive and false-negative rates (53).
Acridine orange stains have a sensitivity of 104 CFU/mL, but require a
fluorescent microscope for visualization (55,56). While there is an increased rate
of detection, the cost and time required have precluded any further interest in this
approach.
2. Bacterial Culture
Because only 0.11 mL of product is typically cultured on an agar plate, the
sensitivity of this method is limited. This may be overcome in part by the use of
automated blood culture systems (57,58). This procedure generally requires
greater than 24 hours for the culture to grow and often is fraught with false
positives resulting from inoculation/culture technique.
3. Endotoxin Assays
The Limulus amebocyte lysate (LAL) assay is the most sensitive, detecting 104
mg of endotoxin per 0.1 mL of plasma, or 104 CFU/mL (59). However, this test
is not widely available and is of limited value; gram-positive bacteria are not
detected by this method because they do not contain endotoxin in their cell walls.
Figure 1 The rRNA chemiluminescence linked probe assay is schematically represented in four stages. (1)
RNA preparation: bacteria are enzymatically lysed and the rRNA is released. (2) Hybridization: a labeled DNA
probe combines with the complementary rRNA to form a stable DNA:RNA hybrid. (3) Differential hydrolysis:
the unhybridized probe is selectively hydrolyzed by base. (4) Detection: a chemiluminescence reaction is
Spinelli and Brecher
initiated in the presence of base and hydrogen peroxide. The amount of luminesence is quantitated in relative
light units (RLU). (From Ref. 23.)
Bacterial Contamination 229
6. Altered Biochemistry
Bacterial metabolism during storage alters certain biochemical properties, namely
glucose, pCO2, pO2, and pH (6467). As bacteria proliferate, they consume
oxygen and glucose, causing a decrease in these analytes, and CO2 is transiently
increased (Fig. 2). Changes in glucose can be rapidly and inexpensively detected
by the use of glucometry (Fig. 3) (65). Two groups have studied the utilization of
dipstick reagents to detect decreased glucose and pH in contaminated platelet
concentrates (64,65). In platelets contaminated with 107 CFU/mL, the overall
sensitivity and specificity were 94% and 98%, respectively (65). In some cases,
S. aureus and K. pneumoniae were detected in the 103105 CFU/mL range (65).
Change in pH has been detected using a CO2-sensitive label placed on the outside
surface of platelet containers. During aerobic growth of bacteria, CO2 diffuses
through the wall of the container and the color in the label changes. Unfortunately,
platelets themselves release CO2 in relation to their number and metabolism,
limiting the utility of this method (68). An alternative technique is the use of
CO2-sensitive labels on gas-permeable plastic bags with culture medium, trans-
ferring only part of the platelet component into it. Unfortunately, this procedure
is also quite insensitive, requiring large numbers of bacteria for detection (69).
7. Visual Inspection
An important observation made by Kim et al. was that grossly contaminated
addition solution (AS) red cell units were darker in appearance than their attached
segmented tubing. As a result of bacterial growth and metabolism, red cells are
230 Spinelli and Brecher
Figure 2 Growth of Yersinia enterocolitica in AS red cells. (a) Growth curves of two
inoculated units indicated by dashed lines. (b) Oxygen tension plotted over storage time
for two sterile (culture negative) and the two inoculated (culture positive) AS3 red cell
units. (c) CO2 tension plotted over storage time for two sterile and the two inoculated AS3
red cell units. (d) Free supernatant hemoglobin over storage time for the same two sterile
and two inoculated AS red cell units. (Figures 2a, b, and d modified from Ref. 23.)
Bacterial Contamination 231
232 Spinelli and Brecher
Figure 3 (a) Glucose analysis of sterile platelets stored in PL-732 bags (mean, mean
-2SD, and mean -3SD) versus platelets stored red in PL-732 bags with CPD and inoculated
with S. aureus (day 0). (b) Glucose analysis of sterile platelets stored in CLX bags (mean,
mean -2SD, and mean-3SD) versus platelets stored in CLX bags with CP2D (double
dextrose) and inoculated with S. marcescens (day 0).
Bacterial Contamination 233
B. Prevention
Many methods have been proposed to aid in the prevention of bacterial contam-
ination of blood products. While many show great promise, there has not been
wide acceptance and institution of any one technique.
1. Donor Questioning
Most cases of red cell contamination are thought to be attributed to transient
asymptomatic bacteremia in donors. Questions are asked prior to donation to
tease out prospective bacteremic donors (24,82). Histories of recent dental
procedures, gastrointestinal or genitourinary manipulation, and breast feeding
may all be associated with bacteremia and would cause a potential donor to be
deferred. Some have suggested asking donors about a recent history of diarrhea
234 Spinelli and Brecher
3. Prestorage Culture
Prestorage culture as a preventive measure against the transfusion of bacteri-
ally contaminated products is problematic. It has been found that 0.20.5%
of units are culture positive (57). As a result of artifactual contamination dur-
ing culturing, the rate of positive cultures may be higher than the amount
of bacterial contamination actually present in stored blood products. A high
number of false positives would likely result in the unnecessary disposal of
noncontaminated units. Additionally, as numerous studies in our and other
laboratories have demonstrated, even with known bacterially inoculated units,
units are frequently culture negative for several days before later cultures be-
come positive (24,90,91). This is likely due to the antibacterial effects of plasma
and granulocytes. Alternatively, breakdown of granulocytes or skin plugs may
lead to a late release of bacteria; thus, many cases may not be detected by
prestorage culture.
4. Bacteriocidal Techniques
The use of antibiotics to assure a sterile product, while effective, is not felt to be
an acceptable solution. Fears of the selection and development of antibiotic-
Bacterial Contamination 235
resistant strains of bacteria and of merely trading one rare event (drug anaphy-
laxis) for another (bacterial sepsis) preclude such use (37,51). A promising
approach is the use of light or ionizing radiation to produce not only non-
immunogenic, but also sterile, blood products (9295). A photochemical decon-
tamination system for platelet concentrates using long-wavelength ultraviolet
radiation and 8-methoxypsoralen has been shown to inactivate 2530 logs/h of
E. coli or S. aureus (96). The techniques that utilize light are restricted to
nonopaque products such as plasma and possibly platelets. With regard to plate-
lets, there has been concern as to the effects this technology might have on
metabolic and functional properties as well as concerns of mutagenicity, logistics,
and cost (97). Because of such concerns and technical hurdles, such methodology
has, to date, been limited to the research laboratory.
5. Storage Time
In 1991, the Blood Product Advisory Committee (BPAC) of FDA reviewed
all cases of Yersinia sepsis from red cell units reported to either FDA or the
CDC during the late 1980s. At that time all reported cases of red cellassociated
yersinia sepsis in the United States had occurred in units older than 25 days.
As a result of the time necessary for the bacteria to attain a lethal concentration,
the BPAC proposed reducing the storage time of red cells from 42 to 25 days (98).
This recommendation was subsequently rejected for the following reasons:
1. A questionnaire distributed at the time revealed that 20% of red cell
units in stock at over 1500 blood banks and transfusion services were
more than 28 days old (98). Discarding such units would have severely
compromised the nations blood supply.
2. A shorter outdate would then require recruitment of new donors; it
was estimated that the addition of a quarter of a million donations
per year would be required to replace the losses due to outdates
(98,99). This would involve additional risk since first-time donors are
known to be at a greater risk of carrying disease because their blood
has not been repeatedly tested, as has the blood of veteran donors
(98,99).
3. Units less than 25 days old can also cause sepsis. While decreasing the
storage time may lessen the problem, it would not eliminate it entirely
(29,33,59,100).
4. Older units are less likely to transmit viruses such as HIV (101).
Increased platelet storage time has also been associated with increased
probability of contamination. At present, the allowable storage time for platelets
is 5 days, although in 1983 the shelf life of platelets was increased to 7 days
(Fig. 4). This increase was associated with an increase in the number of sep-
236 Spinelli and Brecher
tic events in older platelets, and the shelf life was again reduced to 5 days
in 1986 (102). Unfortunately, merely decreasing the shelf life did not elimi-
nate the problem of bacterial contamination. We and other laboratories have
shown that even a moderate inoculate (1050 CFU/mL) of certain bacteria, such
as B. cereus and P. aeruginosa, can have a minimal lag phase with a doubling time
of 12 hours (60). This can lead to a bacterial load of 108 CFU/mL in just 12
days. One approach to minimize platelet associated sepsis taken by the Oncology
Center of Johns Hopkins was to limit the transfusion of platelets to 4 days of
storage (103). It is unlikely that there will be further national reductions in storage
time of platelets because this would cause severe shortages of this component.
Other concerns within the blood banking community are also affecting
platelet availability. Because of the increased complexity of viral screening of
blood products, an increasing awareness of the potential for errors, and a need to
minimize cost and maximize efficiency of operations, many blood collection
organizations such as the American Red Cross are attempting to centralize or
regionalize disease marker testing. While commendable in concept, this central-
ization of testing can, because of the transport time of samples, result in the
decreased availability of one-day-old platelets. In 1982, the mean age of distrib-
uted platelets was 1.6 days, in 1983 (after extension of the dating period to 5 days)
it was 2.0 days, and in 1992 (after addition of increased laboratory testing) it was
2.5 days. In 1983, only 5% of issued platelets were older than 3 days. In 1992,
just 10% were older than 3 days. But with the introduction of centralized testing
by the Red Cross, the mean age of issued platelets increased to 2.7 days, with
20% older than 3 days (89). Not only can this decrease the available shelf life of
an already precariously limited supply of platelets, it can also decrease the
availability of fresh platelets, the most hemostatically effective and the least likely
to be bacterially contaminated.
placed immediately at 4C, growth occurs more rapidly than if held at 10C for
24 hours prior to 4C storage (107). Bacterial growth of most pathogens is
inhibited by cold temperatures, but host defense mechanisms present in fresh
whole blood are also inhibited by storage at 4C. Yersinia with plasmid-encoded
complement resistance can become complement sensitive when blood is transiently
stored at 20C (105). Because phagocytosis and complement activation are impaired
by storage at 4C, it may be advantageous to allow red blood cells to remain in contact
with plasma for several hours prior to separation and storage of components.
Platelets are stored for a maximum of 5 days at 2024C because of concern
regarding the potential for bacterial contamination and progressive decline in
platelet function (storage lesion) if platelets are stored for longer periods of time
at these temperatures (108,109). While storage of platelets at 4C results in a
significantly lower rate of bacterial contamination, it also causes a temperature-
induced activation of platelets and a rapid decline in functional ability and in vitro
viability. In a recent study, cold storage of platelets was performed, mimicking
the endogenous inhibition of platelet activation through the addition of specific
second-messenger stimulators. It was reported that the platelets stored at 4C for
9 days displayed no loss in cell number, and that the study platelets recovered
partial functional ability and viability compared with control platelets stored at
22C for 5 days (110). If a practical method for storing platelets at 4C is
perfected, it would have the potential to reduce the risk of bacterial contamination
of this component.
7. Leukoreduction
Because of concern regarding the accumulation of leukocyte and platelet break-
down products in stored blood, as well as the immunogenicity of white cell
fragments, there has been considerable emphasis placed on the potential benefits
of prestorage versus poststorage leukoreduction (111,112). However, this posed a
potential dilemma, as it was possible that prestorage filtration would cause an
increase in bacterial contamination of blood products and septic complications
because of removal of the phagocytic leukocytes that would normally remove low
levels of bacteria present in these components. Fortunately, in the case of
Y. enterocolitica contamination of red cells, prestorage leukoreduction actually is
associated with a decrease in bacterial growth in inoculated red blood cells
(Table 3) (56,104,106,113,114). The mechanism by which leukoreduction re-
moves bacteria is multifactorial. Bacteria that have been phagocytized (Fig. 5)
but not killed are removed with the white blood cells. Alternatively, organisms
may be adsorbed to leukocytes or activated complement, to then be bound
indirectly to the filter, or the bacteria may directly adhere to the filter fibers
(116,117). Studies on leukoreduction of platelets have not shown such promising
results. Unlike the decrease in Y. enterocolitica growth in prestorage leukoreduced
238 Spinelli and Brecher
red cells, neither a positive nor a negative effect on bacterial growth has been
demonstrated in prestorage leukoreduction of platelets (60,118,119).
8. Frozen Components
Contamination of blood components has occurred during the thawing process
(45,46). Use of a plastic overwrap that prevents outlet ports from direct exposure
to water from the warming bath is an easy and effective method to prevent
contamination (120). Disinfectants and frequent changing of water baths have not
been shown to prevent growth of bacterial species (11,37).
Bacterial Contamination 239
9. Immunoassay
The Red Cross is currently developing an immunochromatographic technique
with Binax, Portland, ME (22,89). This method is intended to be applied im-
mediately prior to issuance of the blood component. The goals of this effort are
to detect the 810 bacteria that account for 95% of bacterially infected platelets,
to detect >105 CFU/mL, to be performed in less than 20 minutes, to be simple
and require no special equipment, and to have a distinct yes or no answer. As yet,
it has not entered clinical trials.
Recently a monoclonal antibody (MAb900) has been described that detects
a prokaryotic elongation factor (EF-Tu). EF-Tu is one of the most abundant
proteins in prokaryotes and is not present in eukaryotic cells. As such, it is hoped
that the use of anti EF-Tu may make a very rapid (46 h) and effective screening
tool possible (121).
240 Spinelli and Brecher
Inoculating
concentration Filtered Control
(CFU/mL) growth/total (%) growth/total (%) Filter type Ref.
V. TRANSFUSION-TRANSMITTED SYPHILIS
Treponema pallidum, the agent that causes syphilis, is a thinwalled, motile,
spirochete. This organism cannot be visualized with Gram stain, nor does it grow
on bacteriological media or cell culture, yet it is considered to be a bacterium,
and infection is treated with penicillin. Although it is a bacterium, it is often
treated as a distinct entity, different from other transfusion-transmitted bacterial
infection and is thus addressed in this separate section of the text. Donors infected
with T. pallidum may be asymptomatic with negative serology during periods of
spirochetemia (122). We are only aware of three cases of transfusiontransmitted
syphilis in the past 27 years (123125). While the organism is killed by storage
at 4C, it may live for 15 days at these cold temperatures (126,127). Subse-
quently, a rare posttranfusion infection may be associated with transfusion of a
very fresh unit of red blood cells from a donor who was in the seronegative phase
at the time of donation. There is greater concern with platelet transfusion, since
this component is stored at 2024C, temperatures suitable for growth of this
organism. The cardiolipin test is used in screening donated blood, but this test lacks
sensitivity for detecting an acute infection. Nevertheless, there is an extremely
low rate of transfusiontransmitted syphilis infection for a number of reasons:
1. Many infected donors are screened with donor questioning.
2. Although an insensitive test in the acute post infections setting, the
cardiolipin assay does pick up a number of infected donors.
3. Refrigerator storage results in the death of spirochetes.
4. Most patients receiving platelets are on antibiotics at the time of
transfusion, which would be bactericidal for any transmitted viable
organisms.
Bacterial Contamination 241
VII. CONCLUSION
Bacterial contamination of blood components remains a problem today. While the
bacterial contamination of red cells, fresh frozen plasma, and cryoprecipitate
occur rarely, it is estimated that approximately 1 in 1000 platelet units are
contaminated as a consequence of storage at 2024C. Thus, the risk of receiving
a bacterially contaminated platelet transfusion exceeds the combined risk of
242 Spinelli and Brecher
and clinicians is to assure the safest blood product possible for both today
and tomorrow.
There remains some controversy regarding the actual risk of bacterial
contamination of blood, and no perfect screening or prevention methodology
currently exists. The perfect should not be the enemy of the good, and im-
plementation of partial solutions that have little risk of causing harm should be
encouraged (135). Rapid bacterial screening of platelets will likely be available
in the near future.
REFERENCES
1. Schreiber GB, Busch MP, Kleinman SH, Korelitz. The risk of tranfusion-transmitted
viral infections. N Engl J Med 1996; 334:16851690.
2. Association Bulletin 96-6. Bacterial Contamination of Blood Components. AABB
Faxnet, No. 294, August 1996. American Association of Blood Banks.
3. Dodd RY. Adverse consequences of blood transfusionquantitative risk estimates.
In: Blood Supply Risks, Perceptions, and Prospects for the Future. Bethesda, MD:
American Association of Blood Banks, 1994:124.
4. Devine P, Linden JV, Hoffstadter LK, et al. Blood donor-, apheresis-, and transfu-
sion-related activities: results of the 1991 American Association of Blood Banks
Institutional Membership Questionnaire. Transfusion 1993; 33:770782.
5. Devine P, Postoway N, Hoffstadter L, et al. Blood donation and transfusion
practices: the 1990 American Association of Blood Banks Institutional Membership
Questionnaire. Transfusion 1992; 32:683687.
6. Surgenor DM, Wallace EL, Hao SHS, et al. Collection and transfusion of blood in
the United States, 1982-1988. N Engl J Med 1990; 322:16461651.
7. Barrett BB, Andersen JW, Anderson KC. Strategies for the avoidance of bacterial
contamination of blood components. Transfusion 1993; 33:228233.
8. Halpin TJ, Kilker S, Epstein J, Tourault M. Bacterial contamination of platelet
poolsOhio, 1991. MMWR 1992; 41:36)37.
9. Morro JF, Braine HG, Kickler TS, Ness PM, Dick JD, Fuller AK. Septic reactions
to platelet transfusions. A persistent problem. JAMA 1991; 266:555558.
10. Chiu EKW, Yuien KY, Lie AKW, Liang R, Lau YL, Lee ACW, Kwong YL, Wong
S, Ng MH, Chan TK. A prosective study of symptomatic bacteremia following
platelet transfusion and its management. Transfusion 1994; 34:950954.
11. Wallas CH. Transfusion of blood components contaminated with bacteria. Transfu-
sion transmitted infections. In: Smith DM, Dodd RY, eds. Transfusion Transmitted
Infections. Chicago: ASCP Press, 1991:195206.
12. Goldman M, Blajchman MA. Blood product-associated bacterial sepsis. Transfusion
Med Rev 1991; 5(1):7383.
13. BPAC discusses invalidation of test results, platelet usage. Blood Bank Week 1992;
9:13.
14. Rhame FS, Root RK, MacLowry JD, Dadismon TA, Bennett JV. Salmonella
244 Spinelli and Brecher
55. Arduino MJ, Bland LA, Tipple MA, et al. Growth and endotoxin production of
Yersinia enterocolitica and Enterobacter agglomerans in packed erythrocytes. J Clin
Microbiol 1989; 27:14831485.
56. Kim DM, Brecher ME, Bland LA, et al. Prestorage removal of Yersinia entercolitica
from red cells with white cell-reduction filters. Transfusion 1992; 32:658662.
57. Blajchman MA, Ali AM. Bacteria in the blood supply: an overlooked issue in
transfusion medicine. In: Nance SJ, ed. Blood safety: Current Challenges. Bethesda,
MD: American Association of Blood Banks, 1992:213228.
58. Myhre BA, Nakusako YY, Schott R. A rapid method for determining the sterility of
frozen-reconstituted blood. Transfusion 1977; 17:195202.
59. Arduino MJ, Bland LA, Tipple MA, et al. Growth and endotoxin production of
Yersinia enterocolitica and Enterobacter agglomerans in packed erythrocytes. J Clin
Microbiol 1989; 27:14831485.
60. Brecher NE, Hogan JJ, Boothe G, et al. The use of a chemiluminescence-linked
universal bacterial ribosomal RNA gene probe and blood gas analysis for the rapid
detection of bacterial contamination in white cell reduced and nonreduced platelets.
Transfusion 1993; 33:450457.
61. Brecher ME, Hogan JJ, Boothe G, et al. Platelet bacterial contamination and the use
of a chemiluminescence-linked universal bacterial ribosomal RNA gene probe.
Transfusion 1994; 34:750755.
62. Sullivan A. Machines speed testing of food for bacteria, Wall Street 1996; (July 15):
63. Feng P, Keasler SP, Hill WE. Direct identification of Yersinia enterocolitica in blood
by polymerase chain reaction amplification. Transfusion 1992; 32:850854.
64. Wagner SJ, Robinette D. Evaluation of swirling, pH, and glucose tests for the
detection of bacterial contamination of platelet concentrates. Transfusion 1996;
36:989993.
65. Burstain JM, Brecher ME, Workman K, Foster M, Faber GH, Mair D. Rapid
identification of bacterially contaminated platelets using reagent strips. Transfusion
1997;
66. Myhre BA, Demainew SH, Yoshimori RN, Nelson E, Carmen RA. pH changes
caused by bacterial growth in contaminated platelet concentrates. Ann Clin Lab Sci
1985; 15:509514.
67. Arpi M, Bremmelgaard A, Abel Y, Olsson K, Hansen L. A novel screening method
for the detection of microbial contamination of platelet concentrates (letter). Vox
Sang 1993; 65:335336.
68. Arpi M, Bremmelgaard A, Abel Y, Olsson K, Hansen L. A novel screening method
for the detection of microbial contamination of platelet concentrates. An experimen-
tal pilot study. Vox Sang 1993; 65:335336.
69. Hogman CF, Gong J. Studies of one invasive and two noninvasive methods for
detection of bacterial contamination of platelet concentrates. Vox Sang 1994;
67:351355.
70. Kim DM, Estes TJ, Brecher ME et al. WBC filtration, blood gas analysis and plasma
hemoglobin in Yersinia enterocolitica contaminated red cells. Transfusion 1992;
32:41S.
71. Kim DM, Brecher ME, Bland LA, Estes TJ, Carmen RA, Nelson EJ. Visual
identification of bacterially contaminated red cells. Transfusion 1992; 32:221225.
Bacterial Contamination 247
72. Myre BA, Walker LJ, White ML. Bacteriocidal properties of platelet concentrates.
Transfusion 1974; 14:116123.
73. Murphy S, Kahn RA, Holme S, et al. Improved storage of platelets for transfusion
in a new container. Blood 1982; 60:194200.
74. Holme S, Murphy S. Quantitative measurements of platelet shape by light transmis-
sion studies: application to storage of platelets for transfusion. J Lab Clin Med 1978;
92:5364.
75. Holme S, Murphy S. Platelet storage at 22C for transfusion: interrelationship of
platelet density and size, medium pH, and viability after in vivo infusion. J Lab Clin
Med 1983; 101:161174.
76. Lindberg JE, Slichter SJ, Murphy S, et al. In vitro function and in vivo viability of
stored platelet concentrates. Effect of a secondary plasticizer component of PVC
storage bags. Transfusion 1983; 23:294299.
77. Murphy S, Sayar SN, Gardner FH. Storage of platelet concentrates at 22C. Blood
1970; 35:549557.
78. White JG, Krivit W. Ultrastructural basis for shape change induced in platelets by
chilling. Blood 1967; 30:625635.
79. Murphy S, Gardner FH. Platelet preservation. Effect of storage temperature on
maintenance of platelet viabilitydeleterious effect of refrigerated storage. N Engl
J Med 1969; 280:10941098.
80. Holme S, Vaidja K, Murphy S. Platelet storage at 22C: effect of type of agitation
on morphology, viability, and function in vitro. Blood 1978; 52:425435.
81. Bertolini F, Murphy S. A multicenter inspection of the swirling phenomenon in
platelet concentrates prepared in routine practice. Transfusion 1996; 36:128132.
82. Grossman BJ, Kollins P, Lau PM, et al. Screening blood donors for gastrointestinal
illness: a strategy to eliminate carriers of Yersinia entercolitica. Transfusion 1991;
31:500501.
83. Katz L, MacPherson JL, Zuck TF. Yersinia and blood donation (letter). Transfusion
1992; 32:191.
84. Hoogkamp-Korstanje JA, de Koning J, Samsom JP. Incidence of human infection
with Yersinia enterocolitica serotypes 03, 08 and 09 and the use of indirect
immunofluorescence in diagnosis. J Infect Dis 1986; 153:138141.
85. Marks MI, Pai CH, Lafleur L, Lackman L, Hammerberg O. Yersinia enterocolitica
gastroenteritis: a prospective study of clinical, bacteriologic, and epidemiologic
features. J Pediatr 1980; 96:2631.
86. Mingrone MG, Fantasia M, Figura N, Guglielmetti P. Characteristics of Yersinia
enterocolitica isolated form children with diarrhea in Italy. J Clin Microbiol 1987;
25:13011304.
87. Olthuis H, Putlaert C, Verhagen C, Valk L. Method for removal of contaminating
bacteria during venapuncture (abstract). Int Soc Blood Transf Regional Congress
1995; 77.
88. Figueroa PI, Yoshimori R, Nelson E, Macabasco G, Carmen R. Distribution of
bacteria in fluid passing through an inoculated collection needle (abstr). Transfusion
1995; 35(suppl):11S.
89. Bacterial detection device. Blood Bank Week 1995; (Sept. 29):2.
248 Spinelli and Brecher
90. Stenhouse MAE, Milner LV. Yersinia enterocolitica: a hazard in blood transfusion.
Transfusion 1982; 22:396398.
91. Van Noyen R, Vandepitte J, Wauters G, Selderslaghs R. Yersinia enterocolitica: its
isolation by cold enrichment from patients and healthy subjects. J Clin Pathol 1981;
34:10521056.
92. Lin L, Wiesehahn GP, Morel PA, et al. The use of 8-methoxypsoralen and long-
wavelength ultraviolet radiation for decontamination of platelet concentrates. Blood
1989; 74:515525.
93. Matthews JL, Newman JT, Sogandares-Bernal F, et al. Photodynamic therapy of
viral contaminants with potential for blood banking applications. Transfusion 1988;
28:8185.
94. Pamphilon DH, Corbin SA, Saunders J, Tandy NP. Applications of ultraviolet light
in the preparation of platelet concentrates. Transfusion 1989; 29:379383.
95. Prince AM, Stephan W, Brotman B. B-propiolactone/ultraviolet irradiation; a review
of its effectiveness for inactivation of viruses in blood derivatives. Rev Infect Dis
1983; 5:92107.
96. Lin L, Wiesehahn GP, Morel PA, Corash L. Use of 8-methoxypsoralen and
long-wavelength ultraviolet radiation for decontamination of platelet concentrates.
Blood 1989; 74:517525.
97. Johnson RB, Napychank PA, Murphy S, Snyder EL. In vitro changes in platelet
function and metabolism following increasing doses of ultraviolet-B irradiation.
Transfusion 1993; 33:249255.
98. FDA committee endorses education and research to combat rare bacterial reaction:
rejects operational changes for now. Council Commun Blood Centers (CCBC)
Newslett 1991; (May 10):14.
99. FDA Blood Products Advisory Committee supports educational efforts on Yersinia
enterocolitica. Blood Bank Week 1991; 20:13.
100. Kostreski F. Dont change storage rules, blood bankers warn. CAP Today 1991; 5,
45.
101. Donegan E, Lenes BA, Tomasulo PA, et al. Transmission of HIV-1 by component
type and duration of shelf storage before transfusion (letter). Transfusion 1991;
30:851852.
102. Braine HG, Kickler TS, Charache P, et al. Bacterial sepsis secondary to platelet
transfusion: an adverse effect of extended storage at room temperature. Transfusion
1986; 26:391393.
103. Food and Drug Administration: Blood Products Advisory Committee, Center for
Biologist Evaluation and Research, 36th Meeting, Rockville, MD, May 29, 1992,
Publication No. 92-026824, p. 433.
104. Buchholz DH, AuBuchon JP, Snyder EL, et al. Removal of Yersinia enterocolitica
from AS-1 Red cells. Transfusion 1992; 32:667672.
105. Gibb AP, Martin Km, Davidson GA, et al. Modeling the growth of Yersinia
enterocolitica in donated blood. Transfusion 1004; 34:304310.
106. Pietersz RNI, Reesink HW, Pauw W, et al. Prevention of Yersinia enterocolitica
growth in red blood cell concentrates. Lancet 1992; 340:755756.
107. Pietersz RNI, Reesink HW, Dekker MA, et al. Elimination of Yersinia enterocolitica
Bacterial Contamination 249
I. INTRODUCTION
251
252 Bianco and Rios
Virus type
gag
Precursor p55 p56
Core p24 p26
Matrix p17 p16
Nucleocapsid p9
Nucleocapsid p7
pol
Reverse transcriptase p66, p51 p68, p53
Endonuclease p31 p34
env
Precursor gp160 gp140
Surface gp120 gp105 (125)
Trans-membrane gp41 gp36 (41)
Regulatory
Vif p23
Vpr p15
Tat p14
Rev p19
Vpu p16
Nef p27
diversity in HIV is generated during the reverse transcription of RNA into cDNA.
The HIV reverse transcriptase (RT) lacks 3 exonuclease proofreading activity
allowing the occurrence of errors during the transcription process. The overall
mutation rate is very high, in the order of 3.4 105 per base per replication cycle.
The degree of genetic variation observed in HIV infection is phenomenalup to
20% within an infected individual (14,15). This high degree of diversity is
particularly seen in patients receiving antiprotease therapy, reaching more than
7% by week 60 (16,17). Diversity is observed even within the same tissue. HIV-1
genomes infecting different regions of the brain of one study subject with HIV
encephalitis (HIVE) had a mosaic structure, being assembled from different
combinations of evolutionarily distinct lineages in p17 (gag), pol, individual
hypervariable regions of gp 120 (V1/V2, V3, V4, and V5), and gp41/nef (18).
Antigenic stimulation influences the dynamics of HIV replication, including the
relative expression of different HIV variants (19). Naturally occurring recombi-
nant HIV strains have been found in infected patients in regions of the world
where multiple genotypic variants coexist (20).
HIV Genome
1000 vpu 8000
gag vif tat
nef
5LTR pol vpr env 3LTR
rev
gag precursor
p15
VPR env precursor
p55 p23
HIV and Blood Transfusion
Figure 1 Graphic representation of the HIV genome and respective gene products.
5LTR- gag-pol-vpr-vpu-env-tat-rev-nef-LRT3 HIV-1 gene productsthe gag (group anti-
gen) region encodes for the structural proteins: p17 (matrix), p24 (capsid), and p15 (p7, p9
nucleocapsid). The pol region encodes for the nonstructural proteins protease (physically
part of the pol open reading frame), p31 (integrase), and p66/p51 (heterodimer reverse
transcriptase). The env (envelope) region encodes for the envelope proteins gp120 (exposed
to the external surface of the virus) and for gp41 (envelope trans-membrane). The genes vpr,
255
tat, rev, and nef encode for p15, p14, p19, and p27, respectively, these are regulatory proteins
involved in viral replication. The genes vif and vpu encode for p23 and p16, respectively.
256 Bianco and Rios
lack of CCR5 expression protected persons from infection. Only individuals who
are homozygous for CCR5 are protected, and there is no difference in the rate of
progression to AIDS between infected heterozygous and homozygous wild-type
subjects (29).
VII. PATHOGENY
The evidence demonstrating that HIV-1 and HIV-2 cause AIDS is overwhelming.
This evidence has been contested by a small number of prominent scientists (30),
but this opinion has been totally rejected by others on the basis of availability of
sound scientific data.
Primary HIV infection is followed by a retroviral syndrome that develops
in 4090% of cases (31). Usual symptoms are flu-like and include fever,
fatigue, pharyngitis, weight loss, myalgias, headache, and nausea. About 50% of
the patients present lymphadenopathy and night sweats. These symptoms disap-
pear after a few weeks. During this period, HIV-1 RNA concentrations vary
widely (104106 molecules/mL). Because of the difficulty in identifying the date
of exposure to HIV-1 infection in persons other than transfusion recipients,
studies of the incubation periods for AIDS have been limited. One study based
on a cohort of 84 homosexual and bisexual men showed that the maximum
likelihood estimate for the proportion of infected homosexual men developing
AIDS was 0.99 (90% CI, 0.381). Furthermore, the maximum likelihood estimate
for the mean incubation period for AIDS in homosexual men was 7.8 years (90%
CI, 4.215.0 years), which was close to the estimate of 8.2 years for adults
developing transfusion-associated AIDS (32). European data on transfusion-
associated (TA) AIDS cases reported in 1989 estimated the median incubation
period in adults from 6.5 to 11 years (33).
HIV and Blood Transfusion 259
years after infection (39). By December 1998 there were 8,760 cases of AIDS
among recipients of blood, blood components, or tissue and 5,145 among patients
with hemophilia or other coagulation disorders among the 688,200 cases of AIDS
reported to CDC (24). This number is somewhat smaller than that predicted by
modeling studies suggesting a total of 15,000 eventual cases of AIDS attributable
to infection by blood transfusion prior to July 1985 (40,41). The incidence of
transfusion-associated HIV-1 infection in San Francisco was estimated to have
risen rapidly from the first occurrence in 1978 to a peak in late 1982 of
approximately 1.1% per transfused unit. The decrease after 1982 coincided with
the implementation of high-risk donor deferral measures. It is estimated that,
overall, approximately 2,135 transfusion recipients were infected with HIV-1 in
the San Francisco region alone (37). By December 1998, 39 cases of AIDS had
been reported among recipients of blood that had screened negative for HIV
antibody (transfused after introduction of screening tests in 1985). The number
of transfusion-associated AIDS cases will certainly be affected by the newer HIV
therapies. Accurate data about transmission will not be available until reporting
of HIV infection (instead of AIDS) is required for the entire United States.
One of the most important sources of information about HIV transmission
by transfusion was a repository of approximately 200,000 sera from blood donors
established in late 1984 and early 1985 by the Transfusion Safety Study (TSS),
which was sponsored by the National Heart, Lung, and Blood Institute (NHLBI).
Collections were made in the four metropolitan areas with the highest prevalence
of AIDS prior to the availability of screening tests for antibodies to HIV in March
1985. Retrospective testing of this repository showed an overall anti-HIV-1
prevalence of 16 cases per 10,000 donations (42). The vast majority of individuals
who received transfusion of blood from seropositive donors became positive for
antibodies to HIV, and many developed AIDS. This was documented by extensive
lookback studies. Essentially, a seropositive donation triggered a search for
records of all prior donations made by that donor and notification of recipients
and testing whenever possible. One of the earliest studies identified seropositive
donors among the units transfused to each of 19 patients who developed AIDS
and were seropositive for HIV (43). HIV-1 was isolated from the blood of blood
donors and HIV-positive blood recipients for months after the transfusion event
clear documentation that HIV viremia was persistent (44). In one rather informa-
tive case, blood components harvested from a single, asymptomatic, seropositive
donor were transfused into a group of cancer patients. Of 10 living recipients,
9 had antibodies to the virus. Cultures for HIV were positive in 7 of the 9
seropositive recipients. Six seropositive recipients had developed immunological
and clinical sequelae of HIV infection (45). A similar case of transmission
occurred in a bone marrow transplant recipient (46).
In the study of a cohort of pediatric patients in a large private metropolitan
hospital, of the 775 children identified as having received transfusions during the
HIV and Blood Transfusion 261
project period, 644 (83%) were located, and 443 (69%) were evaluated for HIV-1
infection. Among those evaluated, 33 (7%) had antibody to HIV-1 (47).
XII. TESTING
The history of introduction of HIV screening tests for blood donor screening has
been extensively reviewed (69).
A. Surrogate Testing
Prior to the availability of a specific screening test for HIV, there were many
attempts to identify surrogate assays that could assist in donor screening. For in-
stance, some scientists at CDC proposed the use of the antibody to the hepatitis
B core antigen (HBcAb) and the detection of urinary neopterin for identification
of donors at risk (70). Others proposed the use of levels of CD4+ cells for the
screening of blood donors (71). However, a study of the correlation of HBcAb
and CD4+ cell counts with use of CUE (a correlate of risk behavior) suggested
that the contribution of these assays in the absence of a specific screening assay
for HIV would be relatively small (72). In addition, determination of T-helper/
T-suppressor cell ratios, HBcAb, and immune complexes among 18 sets of blood
donors who donated blood to recipients who subsequently developed AIDS could
not distinguish suspected transmitters from controls. None of the assays was as
sensitive and specific as the later developed tests for antibody to HIV (73).
264 Bianco and Rios
B. Specific Tests
The first specific tests for antibodies to HIV became available in March 1985.
Within several weeks, the entire blood supply was being screened using the
first-generation assays. These assays had a number of recognized problems, such
as lack of specificity and known cross-reactivity with antibodies to HLA antigens
that were present on the cell line used to grow the virus. There was so much
concern about lack of specificity that the FDA Memorandum to Blood Establish-
ments published in February 1995 determined that a sample was reactive only
when, after an initial reactive screen, it was repeated in duplicate and one of the
duplicates was reactive. There were no confirmatory assays available, and there
was widespread concern about notifying donors of reactive test results in the
absence of adequate sensitivity and specificity data. Most blood-collecting facil-
ities delayed notification of reactive blood donors until the first quarter of 1987,
when the Western blot confirmatory assay was licensed (74). Despite these
caveats, the early HIV screening assays contributed immensely to the safety of
the blood supply. The development of blood donor screening tests for HIV has
been extensively reviewed in the past (75).
incidence of HIV infection among first-time donors is 2.4 times higher than that
of repeat donors (88). Interestingly, concern over the theoretical possibility of
disease transmission via blood from donors who develop Creutzfeldt-Jakob
disease has led to proposals for the deferral of donors who are 50 years of age or
older. This would require recruitment of a higher number of first-time donors to
replace the deferred individuals. The estimated increase in the risk of infected
units would be 12% for HIV, 21% for HCV, and 22% for HBsAg (89). The po-
tential effects of the introduction of research assays for HIV RNA will be
discussed later in this chapter.
XV. CONCLUSION
The control of HIV transmission by transfusion of blood and blood products is
one of the biggest success stories of science and medicine. We certainly will
benefit from the study of all the efforts applied by many people in many different
areas to address this major issue. The major issue that still remains unresolved
for the transfusion medicine community is how to effectively transmit infor-
mation about the safety of the blood supply instead of the risks associated with
blood transfusions.
REFERENCES
1. Centers for Disease Control and Prevention. Epidemiologic notes and reports:
Pneumocystis carinii pneumonia among persons with hemophilia A. MMWR 1982;
31:365367,
2. Centers for Disease Control and Prevention. Update on acquired immunodeficiency
syndrome (AIDS) among patients with hemophilia A. MMWR 1982; 31:644646,
3. Centers for Disease Control and Prevention. Epidemiologic notes and reports:
possible transfusion-associated acquired immune deficiency syndrome (AIDS)
California. MMWR 1982; 31:652654,
4. Jett JR, Kuritsky JN, Katzmann JA, Homburger HA. Acquired immunodeficiency
syndrome associated with blood-product transfusions. Ann Int Med 1983; 99:621
624,
5. Curran JW, Lawrence DW, Jaffe, H, Kaplan JE, et al. Acquired immuno-deficiency
syndrome (AIDS) associated with transfusions. N Engl J Med 1984; 310:6975,
6. Bove, JR. Transfusion associated AIDSa cause for concern. N Engl J Med 1984;
310:115116,
7. Remis RS, OShaughnessy MV, Tsoukas C, Growe GH, Schechter MT, Palmer RW,
Lawrence DN. HIV transmission to patients with hemophilia by heat-treated,
donor-screened factor concentrate. CMAJ 1990; 142:12471254.
272 Bianco and Rios
8. Dietrich SL, Mosley JW, Lusher JM, Hilgartner MW, Operskalski EA, Habel L,
Aledort LM, Gjerset GF, Koerper MA, Lewis BH, et al. Transmission of human
immunodeficiency virus type 1 by dry-heated clotting factor concentrates. Transfu-
sion Safety Study Group. Vox Sang 1990; 59:129135.
9. Zuck TF, Eyster ME. Blood safety decisions, 1982 to 1986: perceptions and
misconceptions. Transfusion 1996; 36:928931.
10. Bianco C. Protecting the blood supply. Transfus Med Rev 1989; 3 (suppl 1):912,
11. Horowitz B, Prince AM, Hamman J, Watklevicz C. Viral safety of solvent/detergent-
treated blood products. Blood Coagul Fibrinolysis 1994; (suppl 3)S2128.
12. Martin MA. The molecular and biological properties of the human immunodefici-
ency virus. In: Stamatoyannopoloulos G, Neinhuis AW, Majerus PW, Varmus H,
eds. The Molecular Basis of Blood Diseases. 2d ed. Philadelphia: W.B. Saunders
Company, 1994:863908.
13. Fauci AS, Desrosiers RC. Pathogenesis of HIV and SIV. In: Coffin JM, Hughes SH,
Varmus HE, eds. Retroviruses. Plainview, NY: Cold Spring Harbor Laboratory
Press, 1997: 587635.
14. Wain-Hobson S. Running the gamut of retroviral variation. Trends Microbiol 1996;
4:135141.
15. Vartanian JP, Sala M, Henry M, Wain-Hobson S, Meyerhans A. Manganese cations
increase the mutation rate of human immunodeficiency virus type 1 ex vivo. J Gen
Virol 1999; 80:19831986.
16. Berkhout B. HIV-1 evolution under pressure of protease inhibitors: climbing the
stairs of viral fitness. J Biomed Sci 1999; 6:298305.
17. Brown AJ, Korber BT, Condra JH. Associations between amino acids in the
evolution of HIV type 1 protease sequences under indinavir therapy. AIDS Res Hum
Retroviruses 1999; 15:247255.
18. Morris A, Marsden M, Halcrow K, Hughes ES, Brettle RP, Bell JE, Simmonds P.
Mosaic structure of the human immunodeficiency virus type 1 genome infecting
lymphoid cells and the brain: evidence for frequent in vivo recombination events in
the evolution of regional populations. J Virol 1999; 73:87208731.
19. Ostrowski MA, Krakauer DC, Li Y, Justement SJ, Learn G, Ehler LA, Stanley SK,
Nowak M, Fauci AS. Effect of immune activation on the dynamics of human
immunodeficiency virus replication and on the distribution of viral quasispecies.
J Virol 1998; 72:77727784.
20. Burke DS. Recombination in HIV: an important viral evolutionary strategy. Emerg
Infect Dis 1997; 3:253259.
21. De Leys R, Vanderborght B, Vanden Haesevelde M, et al. Isolation and partial
characterization of an unusual human immunodeficiency retrovirus from two per-
sons of West-Central African origin. J Virol 1990; 64:12071216.
22. Centers for Disease Control and Prevention. Identification of HIV-1 Group O
InfectionLos Angeles County, California, 1996. MMWR 1996; 45:561565.
23. van Binsbergen J, Keur W, v d Graaf M, Siebelink A, Jacobs A, de Rijk D, Toonen
J, Zekeng L, Afane Ze E, Gurtler LG. Reactivity of a new HIV-1 group O third
generation A-HIV-1/-2 assay with an unusual HIV-1 seroconversion panel and
HIV-1 group O/group M subtyped samples. J Virol Methods 1997; 69:2937.
HIV and Blood Transfusion 273
24. Centers for Disease Control and Prevention. HIV AIDS Surveillance Report 1998;
10:14.
25. Cao Y, Qin L, Zhang L, Safrit J, Ho DD. Virologic and immunologic characteriza-
tion of long-term survivors of human immunodeficiency virus type 1 infection. N
Engl J Med 1995; 332:201208.
26. Pantaleo G, Menzo S, Vaccarezza M, Graziosi C, Cohen OJ, Demarest JF,
Montefiori D, Orenstein JM, Fox C, Schrager LK, et al. Studies in subjects with
long-term nonprogressive human immunodeficiency virus infection. N Engl J Med
1995; 332:209216.
27. de Roda Husman AM, Koot M, Cornelissen M, Keet IP, Brouwer M, Broersen SM,
Bakker M, Roos MT, Prins M, de Wolf F, Coutinho RA, Miedema F, Goudsmit J,
Schuitemaker H. Association between CCR5 genotype and the clinical course of
HIV-1 infection. Ann Intern Med 1997; 127:882.
28. Quillent C, Oberlin E, Braun J, et al. HIV-1-resistance phenotype conferred by
combination of two separate inherited mutations of CCR5 gene. Lancet 1998;
351:1418.
29. Wilkinson DA, Operskalski EA, Busch MP, Mosley JW, Koup RA. A 32-bp deletion
within the CCR5 locus protects against transmission of parenterally acquired human
immunodeficiency virus but does not affect progression to AIDS-defining illness.
J Infect Dis 1998; 178:11631166.
30. Duesberg P, Rasnick D. The AIDS dilemma: drug diseases blamed on a passenger
virus. Genetica 1998; 104:85132.
31. Quinn TC. Acute primary HIV infection. JAMA 1997; 278:5862.
32. Lui KJ, Darrow WW, Rutherford GW 3d. A model-based estimate of the mean
incubation period for AIDS in homosexual men. Science 1988; 240:13331335.
33. Kalbfleisch JD, Lawless JF. Estimating the incubation time distribution and ex-
pected number of cases of transfusion-associated acquired immune deficiency
syndrome. Transfusion 1989; 29:672676.
34. Mylonakis E, Dickinson BP, Mileno MD, Flanigan T, Schiffman FJ, Mega A, Rich
JD. Persistent parvovirus B19 related anemia of seven years duration in an
HIV-infected patient: complete remission associated with highly active antiretroviral
therapy. Am J Hematol 1999; 60:164166.
35. Palella FJ Jr, Delaney KM, Moorman AC, et al. Declining morbidity and mortality
among patients with advanced human immunodeficiency virus infection. HIV
Outpatient Study Investigators. N Engl J Med 1998; 26 (338):853860.
36. Groopman JE. Impact of transfusion on viral load in human immunodeficiency virus
infection. Semin Hematol 1997; 34 (suppl 2):2733.
37. Busch MP, Young MJ, Samson SM, Mosley JW, Ward JW, Perkins HA. Risk of
human immunodeficiency virus (HIV) transmission by blood transfusions before the
implementation of HIV-1 antibody screening. The Transfusion Safety Study Group.
Transfusion 1991; 31:411.
38. Minamoto GY, Scheinberg DA, Dietz K, Gold JW, Chein N, Gee T, Reich LM,
Hoffer J, Mayer K, Armstrong D, et al. Human immunodeficiency virus infection
in patients with leukemia. Blood 1988; 71:11471149.
39. Ward JW, Bush TJ, Perkins HA, Lieb LE, Allen JR, Goldfinger D, Samson SM,
Pepkowitz SH, Fernando LP, Holland PV, et al W, Bush TJ, Perkins HA, Lieb LE,
274 Bianco and Rios
Allen JR, Goldfinger D, Samson SM, Pepkowitz SH, Fernando LP, Holland PV,
et al. The natural history of transfusion-associated infection with human immuno-
deficiency virus. Factors influencing the rate of progression to disease. N Engl J
Med 1989; 321:947952.
40. Kalbfleisch JD, Lawless JF. Estimating the incubation time distribution and ex-
pected number of cases of transfusion-associated acquired immune deficiency
syndrome. Transfusion 1989; 29:672676.
41. Selik RM, Ward JW, Buehler JW. Trends in transfusion-associated acquired immune
deficiency syndrome in the United States, 1982 through 1991. Transfusion 1993;
33:890893.
42. Kleinman SH, Niland JC, Azen SP, Operskalski EA, Barbosa LH, Chernoff Al,
Edwards VM, Lenes BA, Marshall GJ, Nemo GJ, et al. Prevalence of antibodies to
human immunodeficiency virus type 1 among blood donors prior to screening. The
Transfusion Safety Study/NHLBI Donor Repository. Transfusion 1989; 29:572
580.
43. Jaffe HW, Sarngadharan MG, DeVico AL, Bruch L, Getchell JP, Kalayanaraman
VS, Haverkos HW, Stoneburner RL, Gallo RC, Curran JW. Infection with HTLV-
III/LAV and transfusion-associated acquired immunodeficiency syndrome. Sero-
logic evidence of an association. JAMA 1985; 254:770773.
44. Feorino PM, Jaffe HW, Palmer E, Peterman TA, Francis DP, Kalyanaraman VS,
Weinstein RA, Stoneburner RL, Alexander WJ, Raevsky C, et al. Transfusion-
associated acquired immunodeficiency syndrome. Evidence for persistent infection
in blood donors. N Engl J Med 1985; 312:12931296.
45. Anderson KC, Gorgone BC, Marlink RG, Ferriani R, Essex ME, Benz PM,
Groopman JE. Transfusion-acquired human immunodeficiency virus infection among
immunocompromised persons. Ann Intern Med 1986; 105:519527.
46. Antin JH, Smith BR, Ewenstein BM, Arceci RJ, Lipton JM, Page PL, Rappeport
JM. HTLV-III infection after bone marrow trasplantation. Blood 1986; 67:160163.
47. Lieb LE, Mundy TM, Goldfinger D, Pepkowitz SH, Brunell PA, Caldwell MB,
Ward JW. Unrecognized human immunodeficiency virus type 1 infection in a cohort
of transfused neonates: a retrospective investigation. Pediatrics 1995; 95:717721.
48. Samson S, Busch M, Ward J, Garner J, Salk S, Fernando L, Holland P, Rutherford
G, Benjamin R, Perkins H. Identification of HIV-infected transfusion recipients: the
utility of cross-referencing previous donor records with AIDS case reports. Trans-
fusion 1990; 30:214218.
49. OBrien TR, Busch MP, Donegan E, Ward JW, Wong L, Samson SM, Perkins HA,
Altman R, Stoneburner RL, Holmberg SD J. Heterosexual transmission of human
immunodeficiency virus type 1 from transfusion recipients to their sex partners.
J AIDS 1994; 7:705710.
50. Operskalski EA, Stram DO, Busch MP, Huang W, Harris M, Dietrich SL, Schiff ER,
Donegan E, Mosley JW. Role of viral load in heterosexual transmission of human
immunodeficiency virus type 1 by blood transfusion recipients. Transfusion Safety
Study Group. Am J Epidemiol 1997; 146:655661.
51. Busch MP, Operskalski EA, Mosley JW, Lee TH, Henrard D, Herman S, Sachs DH,
Harris M, Huang W, Stram DO. Factors influencing human immunodeficiency virus
HIV and Blood Transfusion 275
67. Gimble JG, Friedman LI. Effects of oral donor questioning about high-risk behav-
iors for human immunodeficiency virus infection. Transfusion 1992; 32(5):446449.
68. Food and Drug Administration. Memorandum to all registered blood establishments.
Revised recommendations for the prevention of HIV transmission by blood and
blood products. (http//:www.fda.gov/cber/publications).
69. Busch MP. Transfusion associated AIDS. In: Rossi EC, Simon TL, Moss GS, Gould
SA, eds. Principles of Transfusion Medicine Baltimore: Williams & Wilkins, 1996:
699708.
70. Fuchs D, Hausen A, Reibnegger G, Reissigl H, Schonitzer D, Spira T, Wachter H.
Urinary neopterin in the diagnosis of acquired immune deficiency syndrome. Eur J
Clin Microbiol 1984; 3:7071.
71. Galel SA, Lifson JD, Engleman EG. Prevention of AIDS transmission through
screening of the blood supply. Annu Rev Immunol 1995; 13:201227.
72. Pindyck J, Waldman A, Oleszko W, Zang E, Bianco C. Prevalence of viral
antibodies and leukocyte abnormalities among blood donors considering themselves
at risk of exposure to AIDS. Ann NY Acad Sci 1984; 437:472484.
73. McDougal JS, Jaffe HW, Cabridilla CD, Sarngadharan MG, Nicholson JK,
Kalyanaraman VS, Schable CA, Kilbourne B, Evatt BL, Gallo RC, et al. Screening
tests for blood donors presumed to have transmitted the acquired immunodeficiency
syndrome. Blood 1985; 65:772775.
74. The Consortium for Retrovirus Serology Standardization. Serological diagnosis of
human immunodeficiency virus infection by Western blot testing. JAMA 1988;
260:675679.
75. Groopman JE, Chen FW, Hope JA, Andrews JM, Swift RL, Benton CV, Sullivan
JL, Volberding PA, Sites DP, Landesman S, et al. Serological characterization of
HTLV-III infection in AIDS and related disorders. J Infect Dis 1986; 153(4):736742.
76. Centers for Disease Control and Prevention. Testing for antibodies to human
immunodeficiency virus type 2 in the United States. MMWR 1992; 41 (RR-no.12).
77. Centers for Disease Control and Prevention. Update: HIV-2 infection among blood
donors and plasma donorsUnited States, June 1992-June 1995. MMWR 1995;
44:603606.
78. Busch MP, Kleinman SH, Williams AE, Smith JW, Ownby HE, Laycock ME, Lee
LL, Pau CP, Schreiber GB. Frequency of human immunodeficiency virus (HIV)
infection among contemporary anti-HIV-1 and anti-HIV-1/2 supplemental test-
indeterminate blood donors. The Retrovirus Epidemiology Donor Study. Transfu-
sion 1996; 36:3744.
79. Kleinman S, Busch MP, Hall L, Thomson R, Glynn S, Gallahan D, Ownby HE,
Williams AE. False-positive HIV-1 test results in a low-risk screening setting of
voluntary blood donation. Retrovirus Epidemiology Donor Study. JAMA 1998;
280:10801085.
80. Ward JW, Holmberg SD, Allen JR, Cohn DL, Critchley SE, Kleinman SH, Lenes
BA, Ravenholt O, Davis JR, Quinn MG, et al. Transmission of human im-
munodeficiency virus (HIV) by blood transfusions screened as negative for HIV
antibody. N Engl J Med 1988; 318:473478.
81. Cohen ND, Munoz A, Reitz BA, Ness PK, Frazier OH, Yawn DH, Lee H, Blattner
W, Donahue JG, Nelson KE, et al. Transmission of retroviruses by transfusion of
HIV and Blood Transfusion 277
95. Korelitz JJ, Busch MP, Williams AE. Antigen testing for human immunodeficiency
virus (HIV) and the magnet effect: Will the benefit of a new HIV test be offset by
the numbers of higher risk, test-seeking donors attracted to blood centers? Retro-
virus Epidemiology Donor Study. Transfusion 1996; 36:203208.
96. Centers for Disease Control and Prevention. U.S. Public Health Service guidelines
for testing and counseling blood and plasma donors for human immunodeficiency
virus type 1 antigen. MMWR 1996; 45 (No. RR-2):19.
97. Hewlett IK, Epstein JS. Food and Drug Administration conference on the feasibility
of genetic technology to close the HIV window in donor screening. Transfusion
1997; 37:346351.
98. Busch MP, Eble BE, Khayam-Bashi H, Heilbron D, Murphy EL, Kwok S, Sninsky
J, Perkins HA, Vyas GN. Evaluation of screened blood donations for human
immunodeficiency virus type 1 infection by culture and DNA amplification of
pooled cells. N Engl J Med 1991; 325:15.
99. Yerly S, Pedrocchi M, Perrin L. The use of polymerase chain reaction in plasma
pools for the concomitant detection of hepatitis C virus and HIV type 1 RNA.
Transfusion 1998; 38:908914.
100. Cardoso MS, Koerner K, Kubanek B. Mini-pool screening by nucleic acid testing
for hepatitis B virus, hepatitis C virus, and HIV: preliminary results. Transfusion
1998; 38:905907.
101. Schottstedt V, Tuma W, Bunger G, Lefevre H. PCR for HBV, HCV and HIV-1
experiences and first results from a routine screening programme in a large blood
transfusion service. Biologicals 1998; 26:101104.
102. Murthy KK, Henrard DR, Eichberg JW, Cobb KE, Busch MP, Allain JP, Alter HJ.
Redefining the HIV-infectious window period in the chimpanzee model: evidence
to suggest that viral nucleic acid testing can prevent blood-borne transmission.
Transfusion 1999; 39:688693.
103. Centers for Disease Control and Prevention. Public Health Service Guidelines for
the Management of Health-Care Worker Exposures to HIV and Recommendations
for Postexposure Prophylaxis. MMWR 1998; 47(RR-7):128.
104. Bianco C, Kessler D. Donor notification and counseling. Management of blood
donors with positive test results. Vox Sang 1994; 67(suppl 3):255259.
105. Emmanuel JC, Bassett MT, Smith HJ, Jacobs JA. Pooling of sera for human
immunodeficiency virus (HIV) testing: an economical method for use in developing
countries. J Clin Pathol 1988; 41:582585.
106. Smillie JM, Ala FA. Reducing the cost of anti-HIV screening. J Virol Methods 1988;
19:181184.
107. AuBuchon JP, Birkmeyer JD, Busch MP. Department of Pathology. Cost-effective-
ness of expanded human immunodeficiency virus-testing protocols for donated
blood. Transfusion 1997; 37:4551.
12
Hepatitis Viruses and
Blood Transfusion
Alfred M. Prince
Lindsley F. Kimball Research Institute, New York Blood Center, New
York, New York
I. HEPATITIS B VIRUS
The classic studies of Krugman at the Willowbrook School were the first to identify
two distinct hepatitis agents, originally termed MS-1 and MS-2, now known as
hepatitis A and hepatitis B virus (1). This made it possible to identify an antigen
discovered in liver tissue in 1967 by immunofluorescence as a hepatitis Bspecific
antigen, (HBsAg) first called the S.H. antigen (2). Blumberg identified an antigen
in the serum of an Australian aborigine that was originally interpreted as a genetic
marker conferring susceptibility to a variety of diseases: leukemia, leprosy, and all
forms of hepatitis (3). However, eventually it became clear that the Australia
antigen was the surface coat of HBV and thus was hepatitis B specific (4).
Numerous studies revealed a statistical asociation between HBsAg and
hepatocellular carcinoma (e.g., Ref. 5). That these reflected an etiological relation-
ship was shown when prospective follow-up of HBsAg carriers and matched
controls revealed that the relative risk of development of hepatocellular carcinoma
was about 224:1 (6), establishing HBV as the leading oncogenic virus in the world.
279
280 Prince
estimated that these would be derived from 47,500 blood donations having both
normal and abnormal ALT levels (22). Only one unit tested positive by PCR, and
even in this case contamination could not be excluded since seroconversion did
not occur and PCR testing was negative on follow-up. If the PCR result is correct,
one positive in 47,500 donations would correspond to 252 PCR positives in the
12 million units of blood transfused in the United States annually. This would be
a higher estimate than the 81 cases preventable by PCR testing given by Schreiber
et al. (21).
all bloodborne non-A, non-B infections in the population studied were likely due
to HCV.
Application of anti-HCV tests rapidly confirmed the importance of these
assays: 80% of cases of chronic posttransfusion hepatitis from different parts of
the world were anti-C100-3 positive (25). Self-limited cases tended to show
transient or no antibody by this assay (31,32). HCV infections are characterized
by a high rate (6080%) of chronic infections. Chronic liver disease in HCV
infections may be manifested as chronic active hepatitis, cirrhosis, and hepato-
cellular carcinoma (33). HCV infections differ in that respect from HBV infec-
tions, where only about 5% of adult infections become chronic.
A strip radio immunoblot assay (RIBA) was manufactured by Chiron
to provide some measure of confirmatory specificity for the ELISA assays.
The concordance between third-generation enzyme immunoassays and the third-
generation immunoblot assay was 96% (34). The third-generation assay is con-
siderably more sensitive than the second-generation assays. Fifty-seven sera with
indeterminate results with the second-generation assay were retested with the
third-generation assay. Thirty-three (57.9%) showed at least one additional band
with the third-generation assay and thus were classified as positive (35).
who had denied drug use during predonation questioning was found to be a major
factor in HCV transmission (38). Intrafamilial transmission of HCV among
spouses has been documented in some (39,40) but not all studies (41). Sex
partners whose spouses were anti-HCV positive were 3.7 times as likely to be
anti-HCV positive as partners of seronegative spouses (42). HCV RNA has been
detected, albeit in very low concentration, in both saliva and urine (43) and rarely
in breast milk (44). Vertical transmission from infected mothers appears to be
extremely rare, except when the mother is coinfected with HIV (45,46). Surgical
intervention, use of nondisposable needles or syringes, and dental therapy have
been implicated in the transmission of HCV (47,48). All of the above possible
routes of transmission need to be evaluated in the analysis of posttransfusion
cases, especially in cases where blood has been screened with second- and third-
generation assays. It should be remembered that over 90% of HCV infections
have been acquired outside the transfusion setting and current testing has reduced
transfusion-transmitted HCV to an extremely low level (33).
E. Course of Hepatitis C
Hepatitis C viremia, as detected by the presence of HCV RNA, occurs within 13
weeks after exposure. About 90% of patients will have detectable anti-HCV 3
months after infection. Chronic hepatitis C is a common clinical syndrome in the
United States and is now the most common reason for liver transplantation. At
least 70% of patients have persistent or intermittent ALT elevations on long-term
follow-up, with liver biopsies showing chronic inflammatory changes. The virus
persists because it continuously evolves into differing variants (quasispecies) as
neutralizing antibodies are developed by the host. About 80% of patients with
chronic HCV will eventually develop cirrhosis and 15% will develop hepato-
cellular carcinoma after 20 years. It is not understood why 15% of those infected
with HCV spontaneously recover or why patients with chronic disease may have
markedly different clinical courses.
Interferon- has been used in the treatment of HCV for several years.
Unfortunately, the treatment results in a sustained response in only 1020% of
patients. Almost all patients who receive interferon experience an uncomfortable
flu-like syndrome early in the course of treatment. More recently, interferon-
and the antiviral agent ribavirin have been given as combined therapy. Sustained
response rates have approximately doubled (to 4050%) using this combination.
There is some controversy about which patients should be treated. However, most
clinicians agree that patients with a persistently elevated ALT, positive HCV
RNA, and a liver biopsy showing portal or bridging fibrosis should be treated.
was also drawn from the fact that no homology to the TTV sequence was found
among 1,731,752 DNA sequences and 154,072 protein sequences in the National
Institute of Genetics (Mishima, Japan) database.
TTV DNA was detected in three of five patients with non-A to G post-
transfusion hepatitis. This virus may belong to the parvovirus group. Its role in
posttransfusion hepatitis deserves further study. The data so far presented suggest
that TTV is an extremely common infection, and that it is not associated with
acute or chronic hepatitis.
To what extent will PCR tests, shortly to be introduced, prevent all residual
posttransfusion hepatitis? This important question will doubtless be clarified
in the next few years. It is not unlikely that posttransfusion hepatitis will soon
be eliminated. Reviews such as the present will then, at best be of only histori-
cal interest.
REFERENCES
1. Krugman S, Giles JP, Hammond J. Infectious hepatitis. Evidence for two distinc-
tive clinical, epidemiological and immunological types of infection. JAMA 1967;
200:365373.
2. Prince AM, Fiji H, Gershon RK. Immunohistochemical studies on the etiology of
anicteric hepatitis in Korea. Am J Hyg 1964; 79:365381.
3. Blumberg BS, Gerstley BJS, Hungerford DA, London WT Sutnick AI. A serum
antigen (Australia antigen) in Downs syndrome, leukemia and hepatitis. Ann Int Med
1967; 66:924931.
4. Prince AM. An antigen detected in the blood during the incubation period of serum
hepatitis Proc Natl Acad Sci USA 1968; 60:814821.
5. Prince AM, Szmuness W, Michon J, Demaille J, Diebolt G, Linhard J, Quenum C,
Sankale M. A case/control study of the association between primary liver cancer and
hepatitis B infection in Senegal. Int J Cancer 1975; 16:376383.
6. Beasley RP, Lin CC, Hwang L-Y, Chien C-S. Risk of hepatocellular carcinoma in
hepatitis B infection: a prospective study in Taiwan. In: Szmuness W, Alter HJ,
Maynard JE, eds. Viral Hepatitis. 1981 International Symposium. Franklin Institute
Press, 1982:261272.
7. Szmuness W, Much MI, Prince AM, Hoofnagle JH, Cherubin CE, Harley EJ, Block
GH. On the role of sexual behavior in the spread of hepatitis B infection. Ann Int
Med 1975; 83:489495.
8. Scott RM, Snitbhan R, Bancroft WH, Alter HJ, Tingpalapong M. Experimental
transmission of hepatitis B virus by semen and saliva. J Infec Dis 1980; 142:6771.
9. Alter HJ, Purcell RH, Gerin JL, London WT, Kaplan PM, McAuliffe WJ, Wagner J,
Holland PV. Transmission of hepatitis B to chimpanzees by hepatitis B surface
antigen-positive saliva and semen. Infect Immun 1977; 16:928933.
10. Davis LG, Weber DJ, Lemon SM. Horizontal transmission of hepatitis B virus.
Lancet 1989; i:889893.
11. Prince AM, White T, Pollock N, Riddle N, Brotman B, Richardson L. The epidemi-
ology of hepatitis B infections in Liberian infants. Infect Immun 1981; 32:675680.
12. Breuer B, Friedman SM, Millner ES, Kane MA, Synder RH, Maynard JE. Transmis-
sion of hepatitis B virus to classroom contacts of mentally retarded carriers. JAMA
1985; 254:31903195.
13. Holland PV, Alter HJ, Purcell RH, Walsh JH, Morrow AG. The infectivity of blood
containing the hepatitis associated antigen. In: Prier JE, Freidman H, eds. Australia
Antigen. Baltimore: University Park Press, 1973:191203.
290 Prince
14. Prince AM, Brotman B, Jass D, Ikram H. Specificity of the direct radioimmunoassay
for the detection of hepatitis B antigen. Lancet 1973; i:13461350.
15. Hoofnagle JH, Seeff LB, Bales ZB, Zimmerman HJ, Veterans Administration Hepa-
titis Cooperative Study Group. Type B hepatitis after transfusion with blood contain-
ing antibody to hepatitis B core antigen. N Engl J Med 1978; 298:13791383.
16. Chung HT, Lee JSK, Lok ASF. Prevention of posttransfusion hepatitis B and C by
screening for antibody to hepatitis C virus and antibody to HBcAg. Hepatology 1993;
18:10451049.
17. Rehermann B, Ferrari C, Pasqunelli C, Chisari FV. The hepatitis B virus persists for
decades after recovery from acute viral hepatitis despite active maintenance of a
cytotoxic T lymphocyte response. Nature Med 1996; 2:11041108.
18. Kline WE, Bowmann RJ, McCurdy KKE, OMalley JP, Sandler SG. Hepatitis B core
antibody (anti-HBc) in blood donors in the United States: implications for surrogate
testing programs. Transfusion 1987; 27:99102.
19. Infectious disease testing for blood transfusions. Consensus development conference
statement. National Institutes of Health, June 911, 1995.
20. Hanson MR, Polesky HF. Evaluation of routine anti-HBc screening of volunteer
blood donors: a questionable surrogate test for non-A, non-B hepatitis. Transfusion
1987; 27:107108.
21. Schreiber GB, Busch MP, Kleinman SH, Korelitz JJ. The risk of transfusion-
transmitted viral infections. N Engl J Med 1996; 334:16851690.
22. Sankary TM, Yang G, Romeo JM, Ulrich PP, Busch MP, Rawal BD, Vyas GN. Rare
detection of hepatitis B and hepatitis C virus genomes by polymerase chain reaction
in seronegative donors with elevated alanine aminotransferase. Transfusion 1994;
34:656660.
23. Prince AM, Brotman B, Grady GF, Kuhns WJ, Hazzi C, Levin RW, Millian SJ. Long
incubation posttransfusion hepatitis without serological evidence of exposure to
hepatitis B virus. Lancet 1974; ii:241246.
24. Feinstone SM, Kapikian AZ, Purcell RH, Alter HJ, Holland PV. Transfusion associ-
ated hepatitis not due to viral hepatitis type A or B. N Engl J Med 1975; 292:767770.
25. Choo Q-L, Weiner AJ, Overby LR, Kuo G, Houghton M, Bradley DW. Hepatitis C
virus: the major causative agent of viral non-A, non-B hepatitis. Br Med Bull 1990;
46:423441.
26. Gonzalez A, Esteban JI, Madoz P, Viladomiu L, Genesca J, Muniz E, Enriquez J,
Torras X, Hernndez JM, Quer J, Vidal X, Alter HJ, Shih JW, Esteban R, Guardia J.
Efficacy of screening donors for antibodies to the hepatitis C virus to prevent
transfusion-associated hepatitis: final report of a prospective trial. Hepatology 1995;
22:439445.
27. Donahue JG, Munoz A, Ness PM, Brown DE Jr, Yawn DH, McAllister HA Jr, Reitz
BA, Nelson KE. The declining risk of post-transfusion hepatitis C virus infection.
N Engl J Med 1992; 327:369373.
28. Uyttendaele S, Claeys H, Mertens H, Verhaert H, Vermylen C. Evaluation of
third-generation screening and confirmatory assays for HCV antibodies. Vox Sang
1994; 66:122129.
29. Silva AE, Hosein B, Boyle RW, Fang CT, Shindo M Waggoner JG, Hoofnagle JH,
Hepatitis Viruses and Blood Transfusion 291
hepatitis C virus genome in saliva and urine of patients with type C hepatitis. J Med
Virol 1993; 41:120128.
44. Zimmermann R, Perucchini D, Fauchere J-C, Joller-Jemelka H, Geyer M, Huch R,
Huch A. Hepatitis C virus in breast milk. Lancet 1995; 345:928.
45. Roudot-Thoraval F, Pawlotsky J-M, Thiers V, Deforges L, Girollet P-P, Guillot F,
Huraux C, Aumont P, Brechot C, Dhumeaux D. Lack of mother-to-infant transmis-
sion of hepatitis C virus in human immunodeficiency virus-seronegative women: a
prospective study with hepatitis C virus RNA testing. Hepatology 1993; 17:772777.
46. Zanetti AR, Tanzi E, Paccagnini S, Principi N, Pizzocolo G, Caccamo ML, DAmico
E, Cambi G, Vecchi L, Lombardy Study Group on Vertical HCV Transmission.
Mother-to-infant transmission of hepatitis C virus. Lancet 1995; 345:289291.
47. Mele A, Sagliocca L, Manzillo G, Converti F, Amoroso P, Stazi MA, Ferrigno L,
Rapicetta M, Franco E, Adamo B, Palumbo F, Sbreglia C, Pan A, Pasquini P, SEIVA
Collaborating Group. Risk factors for acute non-A, non-B hepatitis and their relation-
ship to antibodies for hepatitis C virus: a case-control study. Am J Public Health 1994;
84:16401643.
48. Chen T-Z, Wu J-C, Yen F-S, Sheng W-Y, Hwang S-J, Huo T-I, Lee S-D. Injection
with nondisposable needles as an important route for transmission of acute commu-
nity-acquired hepatitis C virus infection in Taiwan. J Med Virol 1995; 46:247251.
49. Sugitani M, Inchauspe G, Shindo M, Prince AM. Sensitivity of serological assays for
detection of HCV viremic blood donors. Lancet 1992; 339:10181019.
50. Prince AM, Scheffel JW, Moore B. A search for hepatitis C virus polymerase chain
reaction positive but seronegative subjects among blood donors with elevated ami-
notransferase. Transfusion 1997; 37:211214.
51. Prince AM, Brotman B, Huima T, Pascual D, Jaffery M, Inchauspe G. Immunity in
hepatitis C infection. J Infect Dis 1992; 165:438443.
52. Wyatt CA, Andrus L, Brotman B, Huang F, Lee D-H, Prince AM. Immunity in
chimpanzees chronically infected with hepatitis C virus: role of minor quasispecies
in reinfection. J Virol 1998; In Press.
53. Uchida T, Shimojima M, Gotoh K, Shikata T, Tanaka E, Kiyosawa K. Silent
hepatitis B virus mutants are responsible for non-A, non-B, non-C, non-D, non-E
hepatitis. Microbiol Immunol 1994; 38:281285.
54. Alter HJ, Bradley DW. Non-A, non-B hepatitis unrelated to the hepatitis C virus
(non-ABC) Semin Liver Dis 1995; 15:110120.
55. Simons JN, Pilot-Matias TJ, Leary TP, Dawson GJ, Desai SM, Schlauder GG,
Muerhoff AS, Erker JC, Buijk SL, Chalmers ML, Van Sant CL, Mushahwa IK.
Identification of two flavivirus-like genomes in the GB hepatitis agent. Proc Natl
Acad Sci USA 1995; 92:34013405.
56. Simons JN, Leary TP, Dawson GJ, Pilot-Matias TJ, Muerhoff AS, Schlauder GG,
Desai SM. Isolation of novel virus-like sequences associated with human hepatitis.
Nat Med 1995; 1:564569.
57. Linner J, Wages J, Jr, Zhang-Keck ZY, Fry KE. Molecular cloning and disease
association of hepatitis G virus: a transfusion transmissible agent. Science 1996;
271:505508.
58. Miyakawa Y, Mayumi M. Hepatitis G virusa true hepatitis virus or an accidental
tourist? N Engl J Med 1997; 336:795796.
Hepatitis Viruses and Blood Transfusion 293
59. Alter HJ, Nakatsuji Y, Melpolder J, Wages J, Wesley R, Shih JW-K, Kim JP.
The incidence of transfusion-associated hepatitis G virus infection and its relation to
liver disease. N Engl J Med 1997; 336:747754.
60. Lefrere J-J, Loiseau P, Maury J, Lasserre J, Mariotti M, Ravera N, Lerable J, Lefevre
G, Morand-Joubert L, Girot R. Natural history of GBV-C/hepatitis G virus infection
through the follow-up of GBV-C/hepatitis G virus-infected blood donors and recipi-
ents studied by RNA polymerase chain reaction and anti-E2 serology. Blood 1997;
90:37763780.
61. Alter MJ, Gallagher M, Morris TT, Moyer LA, Meeks EL, Krawczynski K, Kim JP,
Margolis HS, Sentinel Counties Viral Hepatitis Study Team. Acute non-A-E hepatitis
in the United States and the role of hepatitis G virus infection. N Engl J Med 1997;
336:741746.
62. Yoshiba M, Okamoto H, Mishiro S. Detection of the GBV-C hepatitis virus genome
in serum from patients with fulminant hepatitis of unknown aetiology. Lancet 1995;
346:11311132.
63. Sato K, Tanaka T, Okamoto H, Miyakawa Y, Mayumi M. Association of circulating
hepatitis G virus with lipoproteins for a lack of binding with antibodies. Biochem
Biophys Res Commun 1996; 229:719725.
64. Nishizawa T, Okamoto H, Konishi K, Yoshizawa H, Miyakawa Y, Mayumi M.
A novel DNA virus (TTV) associated with elevated transaminase levels in post-
transfusion hepatitis of unknown etiology. Biochem Biophy Res Comm 1997;
241:9297.
65. Mulyanto DR, Suwignya S, Tsauri S, Itoh K, Mizui M, Tsuda F, Okamoto H,
Yoshizawa H, Mishiro. An easy dipstick assay for anti-core antibodies to screen
donors for hepatitis C virus viremia. Vox Sang 1996; 70:229231.
66. Allain JP, Kitchen A, Aloysius S, Reeves I, Petrik J, Barbara JAJ, Williamson LM.
Safety and efficacy of hepatitis C virus antibody screening of blood donors with two
sequential screening assays. Transfusion 1996; 36:401405.
13
Human T-Cell Leukemia Virus
The human T-cell leukemia viruses type I (HTLV-I) and type II (HTLV-II) are
human retroviruses in the oncornavirus family. This family includes the human
immunodeficiency virus (HIV), the bovine leukemia virus (BLV), and the simian
T-cell leukemia virus (STLV) (1). HTLV-I was first isolated from a patient with
cutaneous T-cell lymphoma (2,3). The virus was quickly associated with human
diseases, such as adult T-cell leukemia, leading to the addition of the retrovirus
family to the category of human pathogens (27). HTLV-II was isolated from a
patient with a T-cell variant hairy cell leukemia (8).
Retroviruses are enveloped viruses that need to be transcribed into DNA
and integrated into the host cell genome in order to replicate. The viral particles
carry the enzyme reverse transcriptase (RT), which transcribes the viral RNA into
complementary DNA (cDNA). The cDNA is subsequently converted into double-
stranded DNA (dsDNA) and integrated into the host cell genome as proviral DNA
(9). The genomic material of retroviruses is composed of three major regions: gag
(group antigen), which encodes for structural proteins; pol, which encodes for
nonstructural proteins, such as polymerase and proteases; and env, which encodes
for envelope proteins. HTLV-I and HTLV-II also have a set of genes named
tax/rex that encode for proteins that regulate viral replication. The genomic
sequence of the virus is flanked by two long terminal repeats (LTR) with elements
that regulate transcription and join the host cell genome after integration (Fig. 1).
The tax protein increases the rate of transcription initiation by acting on the
promoter located in the 5LTR. The tax protein also transactivates heterologous
295
296
HTLV Genome
1000 8000
p27/p26 rex
Polymerase
Figure 1 Graphic representation of the HTLV genome and respective gene products.
5LTR-gag-pol-env-tax/rex-LRT3HTLV gene productsthe gag (group antigen) region
encodes for the following structural proteins: p19 (matrix), p24 (capsid), and p15 (nu-
cleocapsid). The pol region encodes for the nonstructural proteins protease and reverse
transcriptase. The env (envelope) region encodes for the envelope proteins gp46 (exposed to
the external surface of the virus) and for p21 (envelope transmembrane). The tax/rex region
encodes for p40 (tax) a regulatory, nonstructural protein that regulates activation of viral
transcription by interacting with LTR, and for the p27 (rex) nonstructural protein that
Rios and Bianco
promoters such as the IL-2 and IL-2R promoter, the GM-CSF promoter, the c-fos
and c-cis promoter. The rex protein controls HTLV gene expression at the post
transcriptional level (10,11). Retroviruses infect a wide range of cellular types in
vitro. Fortunately, their ability to transform these cells is limited.
Unlike other retroviruses, HTLV has highly conserved genomic sequences
(12). The rate of base substitutions is estimated at approximately 1% per 1000
years (13,14). The diversity of the entire collection of HTLV sequences identified
to date is lower than that observed within a single patient in late stage of HIV
infection (1417).
II. EPIDEMIOLOGY
HTLV-I is found worldwide. However, it clusters preferentially around the
equatorial belt (Table 1). The prevalence of HTLV-I/II infection in the United
States is approximately 0.0004%, based on blood donor screening. About half of
the positive individuals are infected with HTLV-II (37).
The epidemiology of HTLV-II is not as well known as that of HTLV-I.
Infection is highly prevalent among intravenous drug users (IVDUs) in the United
States and in Europe (4346). Infection with HTLV-II is also frequent among
Native American populations from North, Central, and South America (4556),
and the prevalence can be as high as 2030% (54,57).
The major routes of transmission for HTLV-I and HTLV-II are sexual
contact and from mother to child through breast-feeding, leading to clear familial
clustering of HTLV-I carriers (56,58). HTLV-I infection has also been reported in
children who have not been breast-fed, indicating that other perinatal routes of
transmission have to be considered (60). Vertical transmission (mother-to-child)
occurs with a frequency of 1030% (29,58,61). HTLV-I/II and HIV share the
same routes of infection. However, HTLV-I/II appears to be less infectious than
HIV. This difference has been attributed to viral load in the course of infection
and to the fact that, in the host, HTLV-I always remain cell-associated while
HIV-1 is both cell-associated and cell-free (6264). The rate of infection among
females is higher than that among males (24,29). Studies involving steady
heterosexual couples documented significant concordance of HTLV-I serological
status (65). The efficacy of sexual transmission has been estimated at 53% (44).
Serologically discordant couples were predominantly female positive and male
negative, indicating higher efficiency of transmission from males to females than
from females to males (59,64,65). HTLV-I has been isolated from semen (66).
Transmission from females to males may be associated with other factors, such
as sexual intercourse during menses and bleeding during intercourse (44).
The modes of transmission of HTLV-II are less well documented than that
of HTLV-I. Sexual transmission of HTLV-II has been difficult to study because
298 Rios and Bianco
function, weakness of leg muscles, and sensory disturbances. Spinal cord atrophy,
signs of funicular demyelinization, axonal loss, and gliosis are often found in the
lower thoracic spinal cord. Sometimes the white matter presents lesions in
magnetic resonance imaging.
Analysis of the cerebrospinal fluid shows pleocytosis, high titer of IgG,
and oligoclonality (92). Neuropathologically it appears as a typical immune-
inflammatory disease with infiltration of the parenchyma by CD4+ and CD8+ T
cells. In later stages, the number of inflammatory cells diminishes and they are
almost exclusively CD8+ T-cells. TSP/HAM patients have a very intense im-
munological response to HTLV-I antigens. The relationship between the immune
response and damage to the central nervous system is unclear. There is indication
that HTLV-I proviral load may be a factor, because TSP/HAM patients have 50
times more HTLV-I proviral DNA in peripheral blood lymphocytes than asymp-
tomatic carrier (93). Patients with TSP/HAM show some improvement after
treatment with oral corticosteroids.
C. Infective Dermatitis
Infective dermatitis was described in 1966 in Jamaican children. The association
with HTLV-I infection was made in 1990 by detection of viral genome in skin
biopsies (71). Infective dermatitis is characterized by crusted scabies, corneal
opacities, chronic bronchiectasis, parasitic worm infestation, and progression to
more severe HTLV-Iassociated diseases such as ATL and TSP/HAM (94,95).
D. HTLV-IAssociated Uveitis
HTLV-Iassociated uveitis is characterized by moderate to severe cellular infil-
tration of the vitreous body (vitreous opacity), mild iritis, and moderate retinal
vasculitis (72). Virological and molecular biological findings suggest that cyto-
kines produced by HTLV-Iinfected T cells in the eye play a central role in the
pathogenesis of the disease. The proviral load in blood lymphocytes from patients
with uveitis is higher than that of asymptomatic carriers (72). The inflammation
responds to topical and systemic treatment with corticosteroids and reoccurs in
about 60% of the cases after therapy is discontinued. Some patients present
typical Sjgrens syndrome (9698).
E. Other Diseases
Other diseases observed in HTLV-Iinfected individuals are opportunistic infec-
tions of the lungs, chronic renal insufficiency, lymphadenopathy, arthropathy, and
autoimmune diseases (99101). HTLV-Iassociated infiltrating pneumonitis has
also been reported in some individuals in Japan (100102).
Human T-Cell Leukemia Virus 301
0 10 20 30 40 50 60 70 80 90
% homology
Figure 2 Degree of homology between HTLV-I and HTLV-II at the nucleotide (nt) and
amino acid levels.
304 Rios and Bianco
antibodies to HTLV-I/II. It appears that more than 60% of these samples test
nonreactive in the second assay. Donors with discordant EIA reactivity remain
eligible to donate as long as the screening assay for the next donation tests
nonreactive. The dual EIA strategy facilitates donor counseling by removing
false-positive test results and avoiding indeterminate Western blot test results on
a substantial number of donors.
Amplification of specific viral sequences through the polymerase chain
reaction has been the most productive approach for confirmation of EIA results
and for the discrimination HTLV type (4347, 128,129). Unfortunately, nucleic
acid amplification technologies for HTLV-I/II are not readily available to most
blood-collecting facilities and clinical laboratories.
REFERENCES
1. Poiesz BJ, Sherman M, Saksena N, et al. The biology and epidemiology of the
human T-cell lymphoma/leukemia viruses. In: Neu HC, Levy JA, Weiss RA, eds.
Frontiers of infectious diseases: Focus on HIV London: Churchill Livingston, 1993:
189205.
2. Poiesz BJ, Ruscetti FW, Gazdar AF, Bunn PA, Gallo RC. Detection and isolation of
type C retrovirus particles from fresh and cultured lymphocytes of a patient with
cutaneous T-cell lymphoma. Proc Natl Acad Sci USA 1980; 77:74157419.
3. Yoshida M, Miyoshi I, Hinuma Y. Isolation and characterization of retrovirus from
cell lines of human adult T-cell leukemia and its implication in the disease. Proc
Natl Acad Sci 1984; 79:20312035.
4. Popovic M, Reitz MS, Jr, Sarngadharan MG. The virus of Japanese adult T-cell
leukemia is a member of the human T-cell leukemia/lymphoma virus Group. Nature
1982; 300:6366.
5. Watanabe T, Seiki M, Yoshida M. ATLV (Japanese isolate) and HTLV (US isolate)
are the same strain of retroviruses. Virology 1984; 133:238241.
6. Blattner WA, Kalyanaram VS, Robert-Guroff M, Lister A, Galton DAG, Sarin PS,
Crawford MH, Catovsky D, Greaves M, Gallo R. The human type-C retrovirus,
HTLV, in blacks from the Caribbean region, and relationship to adult T-cell
leukemia/lymphoma. Int J Cancer 1982; 30:257264.
7. Doski H, Denic S, Patel N, Krishnamurthy M, Levine P, Clark JW. Adult T-cell
Leukemia/Lymphoma in Brooklyn. JAMA 1988; 259:22552257.
8. Kalyanaraman VS, Sarngadharan MG, Robert-Guroff M. A new subtype of human
T-cell leukemia virus (HTLV-II) associated with a T-cell variant of hairy cell
leukemia. Science 1982; 218:571573.
9. Weiss R, Teich N, Varmus H, Coffin J, eds. RNA Tumor Viruses. Cold Spring
Harbor, NY: Cold Spring Harbor Laboratory, 1985.
10. Inoue J, Seiki M, Yoshida M. The second pX product p27xIII of HTLV-I is required
for gag gene expression. FEBS 1986; 209:187190.
11. Inoue J, Yoshida M, Seiki M. Transcriptional (p40x) and post-transcriptional
(p27xIII) regulators are required for the expression and replication of human T-cell
leukemia virus type I genes. Proc Natl Sci USA 1987; 84:36533657.
12. Holland J J, ed. Genetic Diversity of RNA Viruses. Berlin: Spring-Verlag, 1992.
13. Sherman MP, Saksena NK, Dube DK, Yanangihara R, Poiesz BJ. Evolutionary
insights in the origin of human T-cell lymphoma/leukemia virus type I (HTLV-I)
derived from sequence analyses of a new HTLV-I variant from Papua New Guinea.
J Virol 1992; 66:25562563.
14. Gessain A, Gallo RC, Franchini G. Low degree of human T-cell leukemia virus type
I genetic drift in vivo as means of monitoring viral transmission and movement of
ancient human populations. J Virol 1992; 66:22882295.
15. Sherman MP, Saksena NK, Dube DK, Yanangihara R, Poiesz BJ. Evolutionary
insights in the origin of human T-cell lymphoma/leukemia virus type I (HTLV-I)
derived from sequence analyses of a new HTLV-I variant from Papua New Guinea.
J Virol 1992; 66:25562563.
306 Rios and Bianco
16. Myers G, Korber B, Wain-Hobson S, Smith RF, Pavlakis GN. Human retroviruses
and AIDS. 1993. Los Alamos, NM:Los Alamos National Laboratory, 1993.
17. Pedrosa Martins L, Chenciner N, Wain-Hobson S. 1992. Complex intra-patient
sequence variation in the V1 and V2 hypervariable regions of the HIV-1 gp120
envelope sequences. Virology 64:837845.
18. Verdier M, Denis F, Sangare A, et al. Prevalence of antibody to human T cell
leukemia virus type I (HTLV-I) in populations of Ivory Coast, West Africa. J Infect
Dis 1989; 160:363370.
19. Touz E, Gessain A, Lyon-Caen O, Gout O. Tropical spastic paraparesis/HTLV-I-
associated myelopathy in Europe and Africa: clinical and epidemiological aspects.
JAIDS Hum Retrov 1996; 13(S1):S3845.
20. Wiktor SZ, Piot P, Mann JM, et al. Human T-cell lymphotropic virus type I (HTLV-I)
among female prostitutes in Kinshasa, Zaire. J Infect Dis 1990; 161:10731077.
21. Blattner WA, Saxinger C, Riedel D. A study of HTLV-I and its associated risk factors
in Trinidad and Tobago. J AIDS 1990; 90:11021108.
22. Blattner WA, Blaney DW, Robert-Guroff M, Sarngadharan MG, Kalyanaraman VS,
Sarin PS, Jaffe ES, Gallo, RC. Epidemiology of human T-cell leukemia/lymphoma
virus. J Infect Dis 1983 147:406416.
23. Riedel D, Evans AS, Saxinger C, Blattner WA. A historical study of human
T-lymphotropic virus type I transmission in Barbados. J Infect Dis 1989; 159:603
609.
24. Murphy EL, Figueroa JP, Gibbs WN, et al. Human T-lymphotropic virus type I
(HTLV-I) seroprevalence in Jamaica: I. Demographic determinants. Am J Epidemiol
1991; 133(11):11141124.
25. Maloney EM, Murphy EL, Figueroa JP, Gibbs WN, Cranston B, Hanchard B,
Holding-Cobham M, Malley K, Blattner WA. Human T-lymphotropic virus type-I
(HTLV-I) seroprevalence in Jamaica II. Geographic and ecological determinants.
Am J Epidemiol 1991; 133:11251134.
26. Taylor GP. The epidemiology of HTLV-I in Europe. JAIDS Hum Retrov 1996;
13(S1):S814.
27. Hale A, Leung T, Sivasubramaniam S, Kenny J, Sutherland S. Prevalence of
antibodies to HTLV in antenatal clinic attendees in south east London. J Med Virol
1997; 52:326329.
28. Voevodin A, Gessain A. Common origin of human T-lymphotropic virus type-I from
Iran, Kuwait, Israel, and La Reunion Island. J Med Virol 1997; 52:7782.
29. Yamaguchi K. Human T-lymphotropic virus type I in Japan. Lancet 1994; 343:213
216.
30. Gabbai AA, Bordin JO, Vieira-Filho JBP, Kuroda A, Oliveira ASB, Cruz MV,
Ribeiro AAF, Delaney SR, Henrard DR, Rosario J, Roman GC. Selectivity of human
T-lymphotropic virus type-I (HTLV-1) and HTLV-2 infection among different
populations in Brasil. Am J Trop Med Hyg 1993; 49:664671.
31. de Rivera IL, Amador L, Mourra S, Li S, Rasheed S. Geographical clustering of
human T-cell lymphotropic virus type 1 infection in Honduras. J Clin Microbiol
1995; 33:29993003.
32. Maloney EM, Ramirez H, Levine A, Blattner WA. A survey of the human T-cell
Human T-Cell Leukemia Virus 307
and injection drug use during pregnancy and vertical transmission of HIV-1. J AIDS
Hum Retrovir 1997; 15:7682.
65. Kaplan JK, Khabbaz RF, Murphy EL, Hermansen S, Roberts C, Lal R, Heneine V,
Wright D, Matijas L, Thomson R, Rudolph D, Switzer WM, Kleinman S, Busch M,
Schreiber GB, and the Retrovirus Epidemiology Donor Study Group. Male-to-
female transmission of human T-cell lymphotropic virus type I and II: association
with viral load. J AIDS 1996; 12:193201.
66. Iwahara Y, Takehara N, Kataoka R, Sawada T, Ohtsuki Y, Nakachi H, Maehama T,
Okayama T, Miyoshi I. Transmission of HTLV-I to rabbits via semen and breast
milk from serositive healthy persons. Int J Cancer 1990; 45:980983.
67. Lal RB, Gongora-Biachi RA, Pardi D, Switzer WM, Goldman I, Lal AA. Evidence
for mother-to-child transmission of human T lymphotropic virus type II. J Infect Dis
1993; 586591.
68. Heneine V, Woods T, Green D, Fukuda K, Giusti R, Castillo L, Armien B, Garcia
F, and Kaplan J E. Detection of HTLV-II in breast milk of HTLV-II infected mothers
(letter). Lancet 1992; 340:11571158.
69. Gessain A, Vernant JC, Maurs L. Antibodies to human T-lymphotropic virus type I
in patients with tropical spastic paraparesis. Lancet 1985; ii:407409.
70. Gessain A, Francis H, Sonan T. HTLV-I and tropical spastic paraparesis in Africa.
Lancet 1986; ii:698.
71. La Grenade L. HTLV-I-associated infective dermatitis: past, present, and future.
J AIDS Hum Retrov 1996; 13(S1):S4649.
72. Mochizuki M, Ono A, Ikeda E, Hikita, Watanabe T, Yamaguchi K, Sagawa K, Ito
K. HTLV-I uveitis. J AIDS Hum Retrov 1996; 13(S1):S5056.
73. Osato M, Yamaguchi K, Yamasaki H, Suzushima H, Asou N, Sakata K, Kawakita
M, Takatsuki K. A human T-cell lymphotropic virus type-I carrier with chronic renal
failure, aplastic anaemia, myelopathy, uveitis, Sjogren syndrome and panniculitis.
Int Med 1996; 35(9):742745.
74. Yamaguchi K, Takatsuki K. Adult T cell leukemia-lymphoma. Baillieres Clin
Hematol 1993; 6:899915.
75. Franchini, G. Molecular mechanisms of human T-cell leukemia/lymphotropic virus
type I infection. Blood 1995; 86:36193639.
76. Blattner WA, Pombo-de-Oliveira, MS. HTLV-I and HTLV-II. In: Merigan C, Bartlett
JG, Bolognesi D, eds. Textbook of AIDS Medicine, 1998: 10031029.
77. Manns A, Hisada M, La Grenade L Human T-lymphotropic virus type I infection.
Lancet 1999; 353:19511958.
78. Yodoi J, Takatsuki K, Masuda T. N Engl J Med 1974; 290:572574.
79. Uchiyama T, Yodoi J, Sagawa, K., Takatsuki K, Uchino, H. Adult T-cell leukemia:
clinical and hematological features of 16 cases. Blood 1977; 50:481492.
80. Hinuma Y, Nagata K, Hanaoka M, et al. Adult T-cell leukemia: antigen in adult
T-cell leukemia cell line and detection of antibodies to the antigen in human sera.
Proc Natl Acad Sci USA 1981; 78:64766480.
81. Yodoi J, Uchiyama T. Diseases associated with HTLV-I: virus, IL-2 receptor
deregulation and redox regulation. Immunol Today 1992; 13:405410.
82. Shimoyama M, members of Lymphoma Study Group. The fourth nationwide study
310 Rios and Bianco
130. Food and Drug Administration (CBER). Guidance for Industry: Donor Screening
for Antibodies to HTLV-II. August 1997
131. Chen YC, Wang CH, Su IJ, Hu CY, Chou MJ, Lee TH, Lin DT, Chung TY, Liu CH,
Yang CS. Infection of human T-cell leukemia virus type I and development of
human T-cell leukemia lymphoma in patients with hematologic neoplasms: a
possible linkage to blood transfusion. Blood 1989; 74(1):388394.
132. Bartholomew C, Jack N, Edwards J, Charles W, Corbin D, Cleghorn FR, Blattner
WA. HTLV-I serostatus of mothers of patients with adult T-cell leukemia and
HTLV-I-associated myelopathy/tropical spastic paraparesis. J Hum Virol 1998;
1(4):302305.
133. Domingues RB, Muniz MR, Jorge ML, Mayo MS, Saez-Alquezar A, Chamone DF,
Scaff M, Marchiori PE. Human T cell lymphotropic virus type-1-associated myelo-
pathy/tropical spastic paraparesis in Sao Paulo, Brazil: association with blood
transfusion. Am J Trop Med Hyg 1997; 57(1):5659.
134. Kuroda Y, Takashima H, Yukitake M, Sakemi T. Development of HTLV-I-associated
myelopathy after blood transfusion in a patient with aplastic anemia and a recipient
of a renal transplant. J Neurol Sci 1992; 109(2):196199.
135. Saxton EH, Lee H, Swanson P, Chen IS, Ruland C, Chin E, Aboulafia D, Delamarter
R, Rosenblatt JD. Detection of human T-cell leukemia/lymphoma virus type I in a
transfusion recipient with chronic myelopathy. Neurology 1989; 89(6):841844.
136. Kaplan JE, Litchfield B, Rouault C, Lairmore MD, Luo CC, Williams L, Brew BJ,
Price RW, Janssen R, Stoneburner R, et al. HTLV-I-associated myelopathy associ-
ated with blood transfusion in the United States: epidemiologic and molecular
evidence linking donor and recipient. Neurology 1991; 41(2 Pt 1):192197.
137. Kurosawa M, Machii T, Kitani T, Tokumine Y, Kawa K, Maekawa I, Kawamura T,
Miyake T, Kanda M. HTLV-I associated myelopathy (HAM) after blood transfusion
in a patient with CD2+ hairy cell leukemia. Am J Clin Pathol 1991; 95(1):7276.
138. Delamarter RB, Carr J, Saxton EH. HTLV-I viral-associated myelopathy after blood
transfusion in a multiple trauma patient. Clin Orthop 1990; (260):191194.
139. Khabbaz RF, Fukuda K, Kaplan JE, Bianco C, Blattner W, Busch M, Dodd R,
Epstein J, Gilcher R, Jackson C, Katz L, Kleinman S, Murphy EL, Nemo G, Poiesz
BJ, Rios M, Sloand E, Sullivan M, William AE. Guidelines for counseling persons
infected with human T-lymphotropic virus type I (HTLV-I) and type II (HTLV-II)
Ann Int Med 1993; 118:448454.
14
Transfusion-Acquired
Cytomegalovirus Infection
Approaching Resolution
Gary E. Tegtmeier
Community Blood Center of Greater Kansas City, Kansas City, Missouri
I. INTRODUCTION
315
316 Tegtmeier
The use of leukoreduced blood products to lower the risk of TA CMV infection
was established subsequently, first in neonates (9), then in BMT patients (10).
Progress in controlling TA CMV infections has been achieved, but an
intractable, low level of risk persists in BMT patients (11). Effective surveillance
for early CMV infections using sensitive laboratory tests such as the pp65
antigenemia test and the availability of effective antiviral agents have reduced
morbidity and mortality in this patient population. Nevertheless, prevention
remains the ultimate goal, one that will certainly be realized in the context of
ongoing research on CMV latency, molecular methods for CMV detection, and
progress in viral inactivation technology.
The aim of this chapter is to review the following areas: (a) recent progress
in understanding the sites of CMV latency and the natural history of CMV
infection as they relate to TA CMV infections; (b) the epidemiology of TA CMV
in recipients; (c) the epidemiology of CMV in donors; and (d) the interventions
currently available to prevent TA CMV infections. The focus will be on the
transmission of CMV by blood components to the seronegative recipient.
If recently infected donors are the source of infection, they may have
low-level viremia and/or larger numbers of latently infected cells than seroposi-
tive donors who were infected at a more remote time. Because leukocytes are not
distributed evenly across different blood components, both the kind and number
of components a patient receives affects the level of risk. Recipient factors that
play a role in transmission are the degree of HLA matching between donor and
recipient, iatrogenic or disease-induced immunosuppression, and the cytokine
environment in the recipient.
Patient populations known to be at risk for severe TA CMV morbidity and
mortality include infants, especially premature infants, pregnant women, solid
organ transplant patients, BMT patients, HIV-infected patients, and patients
with malignancy.
Rates of infection in transfused CMV-seronegative premature infants were
shown to be quite high in early studies, ranging from 24.0 to 31.8% (3,35). Later
investigations failed to corroborate the high rates originally seen; infection rates
varied from 0 to 8.7% (19,3638). No clear explanations for these differences in
rates could be discerned; these studies were carried out in different geographic
regions. The observed differences in rates were not readily correlated with donor
anti-CMV prevalence, the amount or age of the blood transfused, or infant
birth weight.
Seronegative pregnant women undergoing primary CMV infection transmit
the infection with high efficiency to the fetus, often with devastating conse-
quences (39). Although the risk of TA CMV in this patient group has not been
rigorously assessed, a recent study failed to show CMV conversions in 162 CMV-
seronegative pregnant women, 8 of whom were transfused prior to delivery (38).
The risk of TA CMV infections in CMV-seronegative allogeneic BMT
patients receiving marrow from CMV seronegative donors and routine blood
product support has been assessed in three studies. The infection rates ranged
from 31.8 to 50.0% (6,8,10) not dissimilar to the 21.4% rate seen in CMV-
seronegative autologous BMT patients receiving routine blood products after
transplantation (10).
Highly variable levels of risk have been reported in CMV-seronegative
solid organ transplant recipients who received organs from CMV-seronegative
donors, as summarized in Table 1 (4056). An extraordinary number of variables
influencing the potential TA risk combine to account for the heterogeneous results
seen in this setting. The variables include the incidence of CMV infections in the
donor population, the CMV antibody test employed, the age of the transfused
blood, and the methods of blood component preparation.
At the recipient level, the potential of TA CMV infection and disease in
organ transplant patients is influenced by the following factors: the immuno-
suppressive regimen employed; the immunogenicity of the transplanted organs;
320 Tegtmeier
Range of PT CMV
Type of transplant infections (%) Ref.
the first published data suggesting that CMV-seronegative donor blood posed a
reduced risk of CMV transmission.
A large prospective study conducted in 1976 further suggested that CMV-
seronegative blood products posed a lesser risk of CMV transmission than those
from seropositive donors (61). Three of 86 seronegative recipients (3%) receiving
seronegative blood became infected compared to 13 of 54 seronegative recipients
(24%) receiving seropositive blood.
The seminal study demonstrating the efficacy of CMV-seronegative blood
products in preventing neonatal CMV infections appeared in 1981 (3). None of
90 seronegative infants given CMV-seronegative blood products became infected.
In contrast, 10 of 74 (13.5%) seronegative infants given unscreened blood
products (some of which were seropositive) became infected, 5 with serious or
fatal infections. All serious or fatal infections occurred in infants who weighed
less than 1500 g.
Seropositive infants in both the 1971 and 1981 studies showed evidence of
posttransfusion CMV infection at varying frequencies whether they received
CMV-seropositive or seronegative blood. Although it is not known whether the
infections were transfusion acquired or maternally derived, none were symptomatic.
Adding further credibility to the 1981 study from Stanford was a 1983
publication from the Medical College of Virginia, which reported on 178 transfused
neonates (35). Eight (4%) infants became infected, all of whom weighed less than
1050 g. Three of these infants died of CMV-related syndromes, and three others had
symptomatic CMV infections. Infected infants received more blood products than
uninfected infants, a greater percentage of which were CMV antibody positive, and
infected infants were more likely to be born to seronegative mothers.
The combined weight of these studies led to the implementation of donor
screening for antibodies to CMV (anti-CMV) to provide an inventory of CMV-
seronegative blood products with a reduced risk of CMV transmission to sero-
negative low birth weight infants. This became an AABB standard in the
mid-1980s.
It is unclear whether the use of seronegative blood products has been
restricted to seronegative at-risk infants despite a subsequent study from the
Stanford group showing that the use of CMV-seronegative blood products might
place low birth weight infants born to seropositive mothers at risk for developing
CMV disease (62). This was thought to be a consequence of iatrogenic blood loss
with replacement by CMV-seronegative blood, catabolic loss of remaining ma-
ternally acquired antibody, and CMV infection acquired from maternal secretions
during birth or from breast milk.
The impetus for providing CMV-seronegative blood products to CMV-
seronegative BMT recipients with CMV-seronegative marrow donors was gen-
erated by studies from Seattle (6), Glasgow (7), and Minneapolis (8). The
Minnesota and Seattle studies demonstrated that those recipients had greatly
322 Tegtmeier
reduced rates of primary CMV infections compared to recipients who were given
unscreened blood products. The results of these investigations are summarized
in Table 2.
Prevention of TA CMV infection in CMV-seronegative solid organ trans-
plant patients receiving CMV-seronegative organs by the use of CMV-safe
blood products has been recommended (60). Due to the limited transfusion
requirements of renal transplant patients, provision of CMV-seronegative blood
products is feasible. However, the more intensive use of blood in heart, heart-
lung, and liver transplants may outstrip the available CMV-screened inventory.
Fortunately, filtered products can be supplied under these circumstances.
100
Male Female Total
90
80
%Anti-CMV Positive
70
60
50
40
30
20
10
0
1725 2635 3645 4655 5664 >65 Total
Age Ranges (Years)
Figure 1 Prevalence of anti-CMV by gender and age in Kansas City blood donors
from 1992 to 1995. Data from 20,000 donors screened for the first time by enzyme
immunoassay.
Transfusion-Acquired Cytomegalovirus 325
Number (%)
V. LEUKOREDUCTION
A 1977 publication from Duke was the first to suggest leukoreduction as a means
of reducing the risk of TA CMV infections (71). Leukocyte-poor units prepared
by inverted centrifugation of whole blood were given to eight cardiac surgery
patients, one of whom became infected compared to four of six controls who were
given unmanipulated whole blood.
The reduced risk of CMV transmission of frozen deglycerolized red cells
was demonstrated in two studies. One from Boston followed renal dialysis
patients (72). None of 21 seronegative recipients became infected after receiving
frozen blood compared to 3 seronegative recipients who became infected after
receiving conventional blood. One hundred and six seronegative neonates in
Houston were given frozen, deglycerolized red cells, none of which showed
evidence of posttransfusion CMV infection (9).
Later studies employed saline washing to reduce the leukocyte count of red
cell units. Despite this intervention, one published in 1986 reported 6 of 54
neonates (11%) with posttransfusion CMV infection (73). A second reported
seroconversion of one infant of 76 transfused (1.5%) with at least one saline-
washed CMV-seropositive unit (74). No contemporary controls were available
for comparison.
An Australian study was the first to demonstrate the effectiveness of
leukoreduction by filtration (75). Nine of 42 infants (21%) receiving unfiltered
seropositive blood became infected compared to 0 of 30 infants given filtered
blood.
None of 48 premature neonates given filtered blood in three Connecticut
intensive care nurseries became infected with CMV (76). No control population
was followed by these investigators. It should be noted that only the studies from
Duke and Australia included controls.
Six additional studies (10,58,7780) using either filtration and/or centrifu-
gation to leukocyte-reduce red blood cells and platelets given to patients with
326 Tegtmeier
donors with hemoglobin AS (82,83), and adverse recipient reactions have sur-
faced recently, e.g., red-eye syndrome (84).
Since 1997 apheresis platelets that qualify as leukoreduced have been part
of the CMV-safe product mix used to support BMT patients in Seattle. Of 387
patients transplanted during this interval, 4% have experienced primary CMV
infection. It is unclear whether pheresis platelets carry a higher risk of CMV
transmission compared to filtered products, but the question begs to be answered.
Thus, there remains an intractable low level of risk for primary CMV infection
in CMV-seronegative transplant patients who are supported with CMV-safe
blood products (85).
REFERENCES
1. Ho M, Cytomegalovirus: Biology and Infection. 2nd ed. New York: Plenum, 1991.
2. Kaariainen L, Klemola E, Paloheimo J. Rise of cytomegalovirus antibodies in an
infectious mononucleosis-like syndrome after transfusion. Br Med J 1966; 1:12701272.
3. Yeager AS, Grumet FC, Hafleigh EB, Arvin AR, Bradley JS, Prober CG. Prevention
of transfusion-acquired cytomegalovirus infections in newborn infants. J Pediatr
1981; 98:281287.
4. Ho M, Suwansirikul S, Dowling JN, Youngblood LA, Armstrong JA. The trans-
planted kidney as a source of cytomegalovirus infection. N Engl J Med 1975;
293:11091112.
5. Meyers JD, Flournoy N, Thomas ED. Risk factors for cytomegalovirus infection after
human marrow transplantation. J Infect Dis 1986; 153:478488.
6. Bowden RA, Sayers M, Flournoy N, Newton B, Banaji M, Thomas ED, Meyers JD.
Cytomegalovirus immune globulin and seronegative blood products to prevent
primary cytomegalovirus infection after marrow transplantation. N Engl J Med 1986;
314:10061010.
7. Mackinnon S, Burnett AK, Crawford RJ, Cameron S, Leask BG, Sommerville RG.
Transfusion-Acquired Cytomegalovirus 329
39. Stagno S. Cytomegalovirus. In: Remington J, Klein G, eds. Infectious Diseases of the
Fetus and Newborn. Philadelphia: WB Saunders, 1995:312353.
40. Marker SC, Howard RJ, Simmons RL, Kalis JM, Connelly DP, Najarian JS, Balfour
Jr. HM. Cytomegalovirus infection: A quantitative prospective study of three hundred
consecutive renal transplants. Surgery 1981; 89:660671.
41. Harris KR, Saeed AA, Digard NJ, Whiteford K, Geoghegan TA, Lee HA, Slapak M.
Cytomegalovirus titers in kidney transplant donor and recipient: influence of cyclo-
sporine A. Transplant Proc 1984; 16:3133.
42. Rubin RH, Tolkoff-Rubin NE, Oliver R, Rota TR, Hamilton J, Betts RF, Pass RF,
Hillis W, Szmuness W, Farrel ML, Hirsch MS. Multi-center seroepidemiologic study
of the impact of cytomegalovirus infection on renal transplantation. Transplantation
1985; 40:243249.
43. Boyce NW, Hayes K, Gee D, Holdsworth SR, Thomson NM, Scott D, Atkins RC.
Cytomegalovirus infection complicating renal transplantation and its relationship to
acute transplant glomerulopathy. Transplantation 1988; 45:706709.
44. Chou S, Norman DJ. The influence of donor factors other than serologic status on
transmission of cytomegalovirus to transplant recipients. Transplantation 1988;
46:8993.
45. Preiksaitis JK, Rosno S, Grumet C, Merigan TC. Infections due to herpesviruses in
cardiac transplant recipients: role of the donor heart and immunosuppressive therapy.
J Infect Dis 1983; 147:974981.
46. Wreghitt TG, Hakim M, Gray JJ, Kucia S, Wallwork J, English TA. Cytomegalovirus
infections in heart and heart and lung transplant recipients. J Clin Pathol 1988;
41:660667.
47. Dummer JS, Montero CG, Griffith BP, Hardesty RL, Paradis IL, Ho M. Infections in
heart-lung transplant recipients. Transplantation 1986; 41:725729.
48. Wreghitt T. Cytomegalovirus infections in heart and heart-lung transplant recipients.
J Anti-Microb Chem 1989(suppl E); 23:4960.
49. Smyth RL, Scott JP, Borysiewicz LK, Sharples LD, Steart S, Wreghitt T, Gray JJ,
Higenbottam TW, Wallwork J. Cytomegalovirus infection in heart-lung transplant
recipients: risk factors, clinical associations and response to treatment. J Infect Dis
1991; 164:10451050.
50. Singh N, Dummer JS, Kusne S, Breinig MK, Armstrong JA, Makowka L, Starzl TE,
Ho M. Infections with cytomegalovirus and other herpesviruses in 121 liver trans-
plant recipients: transmission by donated organ and the effect of OKT3 antibodies. J
Infect Dis 1988; 158:124131.
51. Barkholt LM, Ericzon B-G, Ehrnst A, Forsgren M, Anderson JP. Cytomegalovirus
infections in liver transplant patients: incidence and outcome. Transplant Proc 1990;
22:235237.
52. Gorensek MJ, Carey WD, Vogt D, Goormastic M. A multivariate analysis of risk
factors for cytomegalovirus infection in liver transplant recipients. Gastroenterology
1990; 98:13261332.
53. Pillay D, Charman H, Burroughs AK, Smith M, Rolles K, Griffiths PD. Surveillance
for CMV infections in orthotopic liver transplant recipients. Transplantation 1992;
53:12611265.
54. Sutherland S, Bracken P, Wreghitt TG, OGrady J, Calne R, Williams R. Donated
332 Tegtmeier
I. INTRODUCTION
335
336 Bianco
III. TSEs
The major human TSEs are kuru, sporadic CJD, iatrogenic CJD, nvCJD, familial
CJD, Gerstmann-Strassler-Scheinker syndrome (GSS), and fatal familial insom-
nia (FFI). Animal TSEs have been known for many years and are useful models
for the human disease. Among them are scrapie in sheep and goats, transmissible
mink encephalopathy (TME), chronic wasting disease of elks, and bovine spongi-
form encephalopathy (BSE), also known as mad cow disease. TSEs appear to
be transmitted by the ingestion of infected tissues from diseased animals. This has
been documented for both TME and BSE (2,3).
A. Kuru
Kuru was the first TSE ever recognized. It occurred among the Fore peoples
inhabiting the Central Highlands of Papua-New Guinea. It was apparently spread
by cannibalism performed as a rite of respect and mourning for the dead. Since
the cessation of cannibalism in the 1950s, the disease has nearly disappeared.
The incubation time exceeded 40 years. The disease was transmitted experimen-
tally to different species of Old and New World monkeys and apes in classic
experiments by Asher, Gibbs, and Gajdusek. Brain contained the highest amount
of infectivity, while serum and blood did not transmit the infection (7). Occasion-
ally, spleen and lymph nodes transmitted disease after intracerebral inoculations,
Human Transmissible Spongiform Encephalopathies 337
but not by administration through a nasogastric tube (four attempts with chim-
panzees) (7). Clinically, Kuru is characterized by a preponderance of cerebel-
lar symptoms; mental deterioration appears later in the course of the disease.
In histological sections, relatively large deposits of PrP (so-called kuru plaques)
can be observed.
B. Sporadic CJD
The sporadic form of CJD is the most common type of human TSE. CJD was first
described in 192021. Most affected individuals die within one year of onset of
symptoms (reviewed in Refs. 4,5). In many cases, a characteristic electroenceph-
alogram with periodic triphasic waves can be observed. The human disease can
be transmitted to nonhuman primates (7) and less regularly to nonprimates (cats,
guinea pigs, mice, and hamsters) (8).
The incidence of CJD has been extensively analyzed by Schonberger et al.
from the CDC. There were 3642 deaths between 1979 and 1994, or 228 deaths/
year, for a rate of about one case per million inhabitants per year in the United
States. The average annual age-adjusted death rate during the study period was
0.95 deaths per million persons, ranging from 0.78 in 1980 to 1.11 in 1987 (9).
The incidence was zero among 5- to 19-year-olds, and reached 3.65.8/1,000,000
among individuals over 60 years of age. The incidence has remained constant
over the years and was the same in all U.S. states. There are no reports of
transmission of CJD by sexual or casual contact. The overall mortality rate in
Europe is 0.69 at one year (10). Interestingly, the phenotypic expression of the
disease may be linked to polymorphism at codon 129 of PrP. In contrast to the
normal population, 69% of CJD patients were homozygous for methionine at
codon 129 (vs. 42% of controls). Homozygosity on codon 129 is considered to
be a genetic predisposing factor for CJD (11).
Clinically, sporadic CJD it is characterized by an early onset of mental
dysfunctions (e.g., memory loss or behavioral abnormalities). However, in a
substantial proportion of cases (15% in a large study of confirmed CJD cases),
cerebellar symptoms are the first signs of the disease (12). The most extensive
experience of experimental transmission of TSEs was reported by Brown et al.
(12) The most effective route of transmission is intracerebral inoculation, and the
highest infectivity is found in the brain of infected individuals, followed by spinal
cord, cerebrospinal fluid, and eye.
C. Iatrogenic CJD
The possibility of iatrogenic transmission of TSEs was first raised in 1974, with
the association of a case of CJD with a corneal transplant performed 18 months
earlier (13). A second case was described more recently with onset of disease 30
338 Bianco
years after a corneal transplant (14). However, these cases are not well docu-
mented, and transmission of CJD by corneal transplants is unlikely. Silver
electrodes used for stereotactic electroencephalograms during neurosurgery trans-
mitted CJD to two patients, although the electrodes had been cleaned and
sterilized by 70% ethanol and formaldehyde vapor between uses (15). No other
transmissions by either of these routes have been reported.
Starting in 1985, CJD was identified among 7 of 6284 recipients of human
pituitary-derived growth hormone prepared from human cadaveric pituitary
glands (16). Concerned about the theoretical possibility of transmission by
transfusion, the U.S. Food and Drug Administration (FDA) recommended as a
precautionary measure that individuals who received human pituitary-derived
growth hormone be deferred from donating blood (November 25, 1987). In
Europe, development of CJD among recipients of human pituitary-derived growth
hormone was also a serious problem. By November 1994, 11 cases had been
reported in the United States, 14 in the United Kingdom, and 32 in France
(L. Schonberger, personal communication). Large batch sizes contributed to the
spread. Duration of the disease was significantly longer than that observed in
sporadic CJD. Estimated mean incubation time was 8.9 years in France and more
than 10 years in the United Kingdom and the United States. Clinical symptoms
consisted mainly of cerebellar ataxia. Homozygosity for codon 129 of the PRNP
gene, the main genetic determinant for susceptibility to CJD, was significantly
associated with disease. The mean incubation time was significantly longer in
heterozygous individuals than in homozygous individuals (11 vs. 8.95 years).
There have been four transmissions traceable to gonadotropin use in Australia.
Gonadotropin was also prepared from cadaveric human pituitary glands but has
had a far more limited use than human growth hormone.
Iatrogenic transmission of CJD has also been documented among recipients
of human dura mater transplants (17). Incubation times were less than 2 years for
some patients. Most, but not all, of the dura mater transmissions have been
associated with use of a single product manufactured in large batches by a single
manufacturer. To date approximately 60 cases have been traced to this source.
Recently, Japan identified 43 additional cases associated with transplants of dura
mater (18). Most of the transmissions occurred in codon 129 homozygous
individuals.
E. Familial TSEs
Three types of CJD, namely familial CJD, GSS (24), and FFI (25), are hereditary
diseases. They are associated with mutations in the normal cellular gene that
codes for the prion protein. GSS evolves very slowly, over a period of years.
Patients with FFI are unable to sleep. Transmission of these TSEs to animals has
been occasionally successful (12).
F. Animal TSEs
Scrapie in sheep has been known for more than 200 years. In 1961, scrapie was
successfully transmitted to laboratory mice (26). Gajdusek and Gibbs were able
to transmit the disease to New and Old World monkeys (reviewed in Refs. 27,28).
In 1975, Manuelidis described the transmission of CJD to guinea pigs by
intracerebral injection of brain tissue (29). These observations were extended to
hamsters, mice, and rats (29,30). The exact route of natural transmission of
scrapie has not been elucidated. Animal experiments are complicated by the
species barrier, i.e., an animal species is less sensitive to a TSE agent isolated
from another species than the originally infected species. In addition, intracereb-
ral injections accept only a limited sample volume (3050 L), raising the
possibility that infectivity is frequently underestimated. Mad cow disease, or
bovine spongiform encephalopathy has been attributed to the practice of feeding
cattle with sheep offal.
26, 1999). The public concern was so intense that it led the European Community
to ban importation of British beef until measures designed to control the spread
of the infection were instituted. The ban was lifted as of August 1,1999. Little is
known about the relative efficiency of various routes of infection by the BSE or
the nvCJD agent. However, the spread of the disease in cattle and its transmission
to other species, presumably through food, suggests that the oral route may be
efficient. In the United States, the Centers for Disease Control and Prevention
(CDC) established surveillance systems for CJD and CJD variants (34). No cases
of BSE have so far been identified. In addition, an analysis of CJD between 1979
and 1994 found no evidence of the variants form of CJD (35). Unfortunately,
because of the very low incidence of CJD and the long incubation period, there
will be a long period before more definitive answers become available.
and probable cases of CJD performed in England and Wales between 198084
and 199092 showed that 21 of the patients had received blood transfusions and
29 had donated blood (55). The frequency of blood transfusions or donations did
not differ between CJD cases and matched controls, leading the investigators to
conclude that the evidence did not suggest that transfusion was a major risk factor
for development of CJD (55).
No cases of CJD among persons with hemophilia had been reported in the
medical literature until October 1994. The Medline database contained 1485
references on CJD and 6385 references on hemophilia between January 1976 and
October 1994. None of these references linked CJD and hemophilia. Unfortu-
nately, this type of search cannot be repeated because of the high number of
articles addressing the theoretical possibility of transmission of CJD by blood
transfusion. An extensive review of mortality data between 1979 and 1994
performed by CDC did not identify a single CJD death in individuals with a
clotting disorder or hemoglobinopathy (35).
A recent multicenter European case control study found no significant risk
of CJD associated with surgery and blood transfusions (56). In a follow-up of one
CJD patient who was a frequent blood donor, none of the blood recipients
developed CJD. Eighteen blood recipients have died of nonneurological disor-
ders; nine were alive at the time of the investigation. The time periods for
follow-up ranged from 1 to 22 years after the blood transfusions.
The distribution of infectivity in plasma derivatives in experimental TSE
models was studied by spiking normal plasma with trypsinized cells from a
scrapie-infected hamster. The plasma was fractionated using the classical Cohn
method and fractions injected intracerebrally into animals. The study showed a
potential but minimal risk of acquiring CJD from the administration of plasma
protein concentrates (57).
E. Leukoreduction
The argument that leukoreduction, a process that removes the vast majority of
leukocytes from blood and blood components, may decrease CJD infectivity of
blood is derived from two different types of observation. First Klein et al. (64)
346 Bianco
immune response, often the basis for a diagnostic test, has not been observed.
Tests indicative of the disease before the onset of clinical symptoms do not
yet exist.
X. OVERALL ASSESSMENT
The assessment of the potential risk of TSE transmission by transfusion has been
a very difficult task. The reality of the AIDS tragedy hit the transfusion medicine
community after years of dismissive statements that minimized risks. In addition,
the hemophilia community, devastated by the transmission of HIV and HCV,
has exerted substantial political pressure, demanding safety and compensation.
Now both the scientific and the blood banking community are afraid of repeating
the same mistakes. The phenomenon could be called the fear of another AIDS
mistake. Medical experts do not want to risk a statement such as there is
sufficient evidence to indicate that the transmission of TSEs by transfusion of
blood and blood products is unlikely out of fear of being proven wrong.
In September 1998 FDA suspended a recall of plasma products that had been
manufactured from pools containing a unit donated by an individual who later
developed classical CJD. They retained recalls for nvCJD and the discard of
components from these donors. These derivative recalls had caused substantial
shortages of plasma derivatives all over the world. However, the fear of another
AIDS mistake prevented the regulators from accepting the overwhelming evi-
dence that individual components also do not transmit classical CJD. Another
interesting observation is the adoption of leukoreduction by several European
countries and Canada despite the lack of epidemiological or experimental evi-
dence that it will contribute to the prevention of CJD transmission. The action
was based on the experiments of Klein et al. (64) suggesting that B cells played
a role in dissemination scrapie in a mouse model quite distant from nvCJD.
However, evidence that blood and plasma have very low infectivity, derived from
the same model, has been summarily ignored.
Despite the evidence against the transmission of classical CJD by transfu-
sion, many of the restrictions, geographic deferrals, recommendations for leuko-
reduction, etc. continue to be applied because the period of follow-up for nvCJD
has been relatively short. The peak of BSE in England occurred in 1990, the first
case of nvCJD was identified in 1996, and as of May 31, 1999, there were 42
definite and probable cases of nvCJD.
The most solid recent assessment has been made by Paul Brown, from the
National Institutes of Health at a meeting in February 1999, Although experi-
mental studies indicate that blood donations from individuals with CJD might be
capable of transmitting disease, the available epidemiological evidence indicates
that bloodborne infection does not occur. He attributed the disparity to (a) very
Human Transmissible Spongiform Encephalopathies 349
low to absent levels of blood infectivity in patients with CJD; (b) dilution
of infectivity in large donor pools; (c) loss of infectivity (13 logs) during
plasma fractionation; and (d) the comparative inefficiency of transmission of the
infectious agent by parenteral routes. Items b and c are only applicable to
plasma derivatives.
ACKNOWLEDGMENTS
The author thanks Maria Rios for assistance in the preparation of this manuscript.
The author also wants to credit the report to the Scientific Committee on
Medicinal Products and Medical Devices of the European Community led by Dr.
J. Lwer, dated October 21, 1998, for its insights into the subject. The report is
posted at the following internet site: http://europa.eu.int/comm/dg24/health/sc/
scmp/outcome_en.html.
REFERENCES
1. Haywood A M. Transmissible spongiform encephalopathies. N Engl J Med 1997;
337:18211828.
2. Johnson RT, Gibbs CJ Jr. Creutzfeldt-Jakob disease and related transmissible spongi-
form encephalopathies. N Engl J Med 1998; 339:19942004.
3. Knight R. The relationship between new variant Creutzfeldt-Jakob disease and bovine
spongiform encephalopathy. Vox Sang 1999; 79:203208.
4. DeArmond SJ, Prusiner SB. Etiology and pathogenesis of prion diseases. AM J
Pathol 1995; 146:785.
5. Gajdusek DC. Nucleation of amyloidogenesis in infectious and non-infectious amy-
loidoses of the brain. Ann NY Acad Sci 1994; 724:173.
6. Manuelidis L. The dimensions of Creutzfeldt-Jakob disease. Transfusion 1994;
34:915.
7. Asher DM, Gibbs CJ Jr, Gajdusek DC. Pathogenesis of subacute spongiform enceph-
alopathies. Ann Clin Lab Sci 1976; 6:84103.
8. Manuelidis EE, Kim JH, Mericangas JR, Manuelidis L. Transmission to animals of
Creutzfeldt-Jakob disease from human blood. Lancet 1985; ii:896.
9. Holman RC, Khan AS Belay ED, Schonberger LB. Creutzfeldt-Jakob disease in the
United States, 1979-1994: using national mortality data to assess the possible
occurrence of variant cases. Emerg Infect Dis 1996; 2:333337.
10. Will RG, Alperovitch A, Poser S, Pocchiari M, Hofman A, Mitrova E, de silva R,
D Alessandro M, Delasnerie-Laupretre N, Zerr I, van Duijn C. Descriptive epidemi-
ology of Creutzfeldt-Jakob disease in six European countries, 1993-1995. Ann Neurol
1998; 43:763767.
11. Parchi P, Castellani R, Capellari S, Ghetti B, Young K, Chen SG, et al. Molecular
basis of phenotypic variability in sporadic Creutzfeldt-Jakob disease. Ann Neurol
1998; 39:767778.
350 Bianco
12. Brown P, Gibbs CJ Jr, Rodgers-Johnson P, Asher DM, Sulima MP, Bacote A,
Goldfarb LG, Gajdusek DC. Human spongiform encephalopathy: the National Insti-
tutes of Health series of 300 cases of experimetally transmitted disease. Ann Neurol
1994; 35:513529.
13. Duffy P, Wolf J, Collins G, DeVoe A G, Steeten B, Cowen D. Possible person-to-
person transmission of Creutzfeldt-Jakob disease. N Engl J Med 1974; 290:692693.
14. Heckmann JG, Lang CJG, Petruch F, Druschky A, Erb C, Brown P, Neundrfer B.
Transmission of Creutzfeldt-Jakob disease via corneal transplant. J Neurol Neurosurg
Psychiatry 1997; 63:388390.
15. Bernoulli C, Siegfried J, Baumgartner G, Regli F, Rabinowicz T, Gajdusek DC, Gibbs
CJ Jr. Danger of accidental person-to-person transmission of Creutzfeldt-Jakob
disease by surgery. Lancet 1977; i:478479.
16. Fradkin JE, Schonberger LB, Mills JL, et al. Creutzfeldt-Jakob disease in pituitary
growth hormone recipients in the United States. JAMA 1991; 265:880.
17. Martinez-Lage JF, Poza M, Sola J, Tortosa JG, Brown P, Cervenakova L, Esteban JA,
Mendonza A. Accidental transmission of Cretzfeldt-Jakob disease by dural cadaveric
grafts. J Neurol Neurosurg Psychiatry 1994; 57:10911094.
18. Centers for Disease Control. Cretzfeldt-Jakob disease associated with cadaveric dura
mater graftsJapan, January 1979-May. MMWR 1996; 46:10661096.
19. Will RG, Ironside JW, Zeidler M, Cousens SN, Estibeiro K, Alperovitch A, Poser S,
Pocchiari M, Hofman A, Smith PG, A new variant of Cretzfeldt-Jakob disease in the
UK. Lancet 1996; 347:921925.
20. Will RG, Knight RSG, Zeidler M, Ironside JW, Cousens SN, et al. Reporting of
suspect new variant Creutzfeldt-Jakob disease. Lancet 1997; 349:847.
21. Hill AF, Butterworth RJ, Joiner D, Rossor MN, Thomas DJ, Frosh A, Tolley N, Bell
JE, Spencer M, King A, AI-Sarraj S, Ironside JW, Iantos PL, Collinge J. Investigation
of variant Cretzfeldt-Jakob disease and other human prion diseases with tonsil biopsy
samples. Lancet 1999; 353:183189.
22. Collinge J, Sidle KCL, Meads J, Ironside J, Hill AF. Molecular analysis of prion strain
variation and the etiology of new variant CJD. Nature 1999; 383:685690.
23. Bruce ME, Will RG, Ironside JW, McConnell I, Drummond D, Suttie A, McCardle
L, Chree A, Hope J, Birkett C, Cousens S, Fraser H, Bostock CJ. Transmissions to
mice indicate that new variant CJD is caused by the BSE agent. Nature 1997;
389:498501.
24. Gerstmann J, Strussler E, Scheinker I. ber eine eigenartige hereditr-familire
Erkrankung des Zentralnervensystems. Zugleich ein Beitrag zur Frage des vor-
zeitigen lokalen Alterns. Z Ges Neurol Psych 1936; 154:736762.
25. Lugaresi E, Medori R, Montagna P, Baruzzi A, Cortelli P, Lugaresi A, et al. Fatal
familial insomnia and dysautonomia with selective degeneration of thalamic nuclei.
N Engl J Med 1986; 315:9971003.
26. Chandler R L. Encephalopathy in mice produced by inoculation with scrapie brain
material. Lancet 1961; i:107108.
27. Gibbs CJ Jr, Gajdusek DC, Morris JA. Viral characteristics of the scrapie agent in
mice. In: Gajdusek DC, Gibbs, CJ, Alper M, eds Slow, Latent and Temperate Virus
Infections. National Institute of Neurologic Diseases and Blindness, 1965:195202.
28. Gibbs CJ Jr, Gajdusek DC. Isolation and characterization of the subacute spongiform
Human Transmissible Spongiform Encephalopathies 351
47. Pattison IH, Millson GC, Smith K. An examination of the action of whole blood,
blood cells or serum on the goat scrapie agent. Res Vet Sci 1964; 5:116121.
48. Marsh RF, Burger D, Hanson RP. Transmissible mink encephalopathy: behavior of
the disease agent in mink. Am J Vet Res 1969; 30:16371642.
49. Marsh RF, Miller JM, Hanson RP. Transmissible mink encephalopathy: studies on
the peripheral lymphocyte. Infect Immun 1973; 7:352355.
50. Hadlow WJ, Eklund CM, Kennedy RC, Jackson TA, Whitford HW, Boyle CC.
Course of experimental scrapie virus infection in the goat. J Infect Dis 1974;
129:559567.
51. Hadlow WJ, Kennedy RC, Race RE, Eklund CM. Virologic and neurohistologic
findings in dairy goats affected with natural scrapie. Vet Pathol 1980; 17:187199.
52. Hadlow WJ, Race RE, Kennedy RC. Temporal distribution of transmissible mink
encephalopathy virus in mink inoculated subcutaneously. J Virol 1987; 61:3235
3240.
53. Wells GAH, Dawson M, Hawkins SAC, Austin AR, Green RB, Dexter I, Horigan
MW, Simmons MM. Preliminary observations on the pathogenesis of experimental
bovine spongiform encephalopathy. In: Gibbs CJ Jr, ed. Bovine Spongiform Enceph-
alopathy: The BSE Dilemma. New York: Springer-Verlag, 1996:2844.
54. Wells GAH, Hawkins SAC, Green RB, Austin AR, Dexter I, Spencer YI, Chaplin
MJ, Stack MJ, Dawson M. Preliminary observations on the pathogenesis of ex-
perimental bovine spongiform encephalopathy (BSE): an update. Vet Rec 1998;
142:103106.
55. Esmonde TFG, Will RG, Slattery JM, Knight R, Harries-Jones R, de Silva R,
Matthews WB. Creutzfeldt-Jakob disease and blood transfusion. Lancet 1993;
341:205207.
56. van Duijn CM, Delasnerie-Lauprtre N, Masullo C, Zerr I, de Silva R, Wientjens
DPWM, Brandel J-P, Weber T, Bonavita V, Zeidler M, Alprovitch A, Poser S,
Granieri E, Heye N, Hensen S, Mller N. Creutzfeldt-Jakob disease and blood
transfusion (letter). Lancet 1994; 343:298299.
57. Brown P, Rohwer RG, Dunstan BC, Macauley C, Gajdusek DC, Drohan WN.
The distribution of infectivity in blood components and plasma derivatives in ex-
perimental models of transmissible spongiform encephalopathy. Transfusion 1998;
38:810816.
58. Sullivan MT, Schonberger LB, Kessler D, et al. Creutzfeldt-Jakob disease (CJD)
investigational lookback study. Transfusion 1997; 37(suppl):2S.
59. Operaskalski EA, Mosley JW. Pooled plasma derivatives and Creutzfeldt-Jakob
disease (letter). Lancet 1995; 346:12241224.
60. Evatt BL. Prions and hemophilia: assessment of risk. Haemophilia 1998; 4:628633.
61. Rahman A, Bullman TA, Kang HK, Muniz A, Kizer KW, Roselle GA, Booss J.
A search for Creutzfeldt-Jakob disease in veterans treated with potentially contami-
nated blood products. Infect Control Hosp Epidemiol. In press.
62. Crange A, Gray F, Cesaro P, Adle-Biassette H, Duvoux C, Cherqui D, Bell J, Parchi
P, Gambetti P, Degos J-D. Creutzfeldt-Jakob disease after liver transplantation. Ann
Neurol 1995; 38:269272.
63. Busch MP, Glynn SA, Schreiber GB. Potential increased risk of virus transmission
Human Transmissible Spongiform Encephalopathies 353
I. INTRODUCTION
II. MALARIA
A. Historical Aspects and Epidemiology
The first case of TTM was reported in 1911 by Wolsey (1), and more than 3000
cases have been described so far (2), although these numbers may represent less
than 50% of the actual cases (3). Although malaria has been eradicated in almost
all European countries, the United States, Australia, and Japan, it is still present
in 102 countries (46), where more than 120 million people are infected annually,
355
356 Wendel
with 1 million deaths and nearly 300 million carrying the parasite. Countries in
tropical Africa are responsible for 80% of all clinical cases and more than 90%
of all parasite carriers in the world. Excluding the African continent, 90% of cases
reported to WHO are from 19 different countries, with some 75% of them
concentrated in 9 countries: India, Brazil, Afghanistan, Sri Lanka, Thailand,
Indonesia, Vietnam, Cambodia, and China (decreasing order of frequency).
Malaria is concentrated in certain regions within a single country. For example,
though 85% of the Brazilian geographic territory is exposed to malaria, less than
15% of the Brazilian population lives in the affected area, with nearly 99% of all
cases detected in the Amazon region. In addition, nearly 40% of the world
population, or 1.8 billion people (1992), still remain exposed to varying degrees
of risk of malarial infection (4,5).
Transmission by blood components is either a consequence of the sanitary
conditions of a country or region, when complete eradication has not yet been
achieved (e.g., tropical Africa, India, Sri Lanka, Brazilian Amazon basin, etc.), or
a result of imported cases into developed, nonendemic places by immigrants or
travelers from endemic areasmainly described in France, the United Kingdom,
the United States, and Spain (311).
The annual incidence of TTM ranges from 0.25 cases/million units in the
United States (12) to more than 50 cases/million units in endemic regions (79).
Even in nonendemic countries where complete eradication has been accom-
plished, the ever-increasing migration flow played a great role in the last three
decades, as observed in France in the 1980s when more than 100 cases were
described (10). Table 1 shows the prevalence of infected donors from some
endemic and nonendemic countries.
%
Infected
Author (Ref.) Country donors Species Method
Chikwem et al. (42) Nigeria 4.0 P.f. Thick and thin blood films
Ibhanesebor et al. (24) Nigeria 40 P.f. Thick and thin blood films
Ferreira et al. (19) Brazila 32 P.f. IgG antibodies (IFAT)
24 P.v.
37 P.f.+P.v.
Kiesslich et al. (20) Brazila 15.7 P.f.+P.v. IFAT
0.8 Thick films
1.6 Acridine orange (QBC)
Hong et al. (41) Vietnam 0.2 P.f. PCR (repetitive genomic DNA)
Chiodini et al. (27) United 1.5b P.f. ELISA (antibody)
Kingdom 0.45c
Tabor (11) France 10b IFAT
P.f. = Plasmodium falciparum; P.v. = Plasmodium vivax; IFAT = indirect immunofluorescence assay;
PCR = polymerase chain reaction.
aDonors only from the Brazilian Amazon, a highly endemic area. Other parts of the country are not
considered.
bDonors from endemic, tropical countries.
cDonors from nonendemic countries, never exposed to malaria. This group most likely represent
ing other red cells and continuing this asexual stage. This schizogonic process is
regular, lasting 48 hours for P. malariae, P. vivax, and P. ovale (Tertian fever) and
72 hours for P. falciparum (Quartan fever).
The merozoites released from liver cells cannot subsequently reinvade other
hepatocytes and perpetuate the process. However, some P. vivax and P. ovale
sporozoites give rise to hypnozoites (a very latent exoerythrocytic form), which
stay dormant inside the hepatic parenchyma for approximately up to 5 years
before starting the exoerythrocytic schizogonic stage, inducing late relapses if
proper treatment is not applied.
In parallel, after several divisions, some erythrocytic parasites undergo
another cycle step and differentiate into sexual gametocytes. They are released in
the bloodstream and, instead of invading red cells, are ingested by another
mosquito during a blood meal, beginning a sexual reproduction (fertilization) in
the insect stomach. This step leads to the development of oocysts under the basal
membrane, which undergo a sporogony (a reduction division), releasing thou-
sands of free sporozoites in the insects hemocele that migrate towards the
mosquito salivary gland, ending their life cycle (6,11,13,14).
358 Wendel
Figure 1 The life cycle of malaria. Although recently infected donors bearing sporozo-
ites in the bloodstream are able to induce infection in experimentally infected volunteers,
this is a very unusual situation. TTM has been observed basically from donors in the
erythrocytic stage through the transfusion of merozoite-infected red cells.
safe if used exclusively for fractionation. Though viability at 46C storage is less
than 5 days for P. malariae and up to 10 days for P. falciparum, holding units for
710 days in order to achieve a relative protection is not justified.
A survey covering the 19731980 period showed distribution of 38% for
P. malariae, 42% for P. vivax, 42% for P. ovale, and 20% for P. falciparum (7).
Another survey conducted in the United States (12) covering the 19721988
period, when 45 TTM cases were reported, showed that a distribution of 38% for
P. malariae, 29% for P. falciparum, 24% for P. vivax, and 9% for P. ovale.
360 Wendel
Conversely, the distribution among different endemic regions also shows a wide
variation; in Africa, P. falciparum is the most common agent (4,5), while P. vivax
is the most common in India (17) and Brazil (1820).
D. Clinical Symptoms
The incubation period depends on several factors: the number of viable transfused
parasites, the species and the strain, the host immunity, and the previous use of
malarial chemoprophylaxis. On average, TTM has a longer incubation period than
that observed in natural transmission, with a 16-day mean period (829 days) for
P. falciparum, a 19.6day mean period (830 days) for P. vivax and P. ovale, and
a 57.2-day mean period (6106 days) for P. malariae (2,7,8).
The first symptoms are usually nonspecific, mainly fever that only reaches
its peculiar periodicity in 2 weeks. Since most physicians in nonendemic areas
are unaware of this possibility, a period is observed between the onset of the
symptoms and diagnosis ranging from 12 to 43 days, but periods of 160 days up
Protozoan Parasites 361
to one year have been reported. Even in endemic countries, a long delay may be
observed (18). Symptoms are more severe in splenectomized, organ-transplanted,
or immunodeficient recipients. In developed countries, TTM is usually associated
with previously splenectomized recipients, cardiopulmonary bypass surgery pa-
tients, and organ transplant recipients (2123), whereas in endemic countries,
particularly in Africa, neonates who undergo exchange transfusions are one of the
most severely affected patient groups (24).
The rate of lethal cases is highly related to late diagnosis (especially in
nonimmune recipients) and plays an important role when infection is due to
P. falciparum (with cerebral, renal, and pulmonary lesions). This was particularly
important in the United States after the Vietnam war when a 24-fold higher
mortality was seen in patients diagnosed in civilian hospitals compared to those
in military hospitals (11). Fatalities are still high, with some reports of up to 20%
of cases, much higher than the imported cases (<1%), where the index of
suspicion is higher. Thus, one must always bear in mind this possibility whenever
fever ensues after a transfusion episode. Since no exoerythrocytic stage is
observed in TTM, no relapse is found (irrespective of the causative species) after
appropriate treatment.
E. Preventive Measures
A dramatic decrease in TTM should be expected after malaria eradication in the
United States. American troops returning from Korea, Vietnam, and, lately,
Somalia brought back a remarkable number of infected personnel responsible for
TTM, despite preventive measures having been taken. On the other hand,
migratory movements, particularly from Africa and India into Europe and from
Southeast Asia and South America into North America (3), were also responsible
for TTM. Therefore, specific measures have been taken in order to prevent it.
The first strategy is taking an accurate history including place of birth,
previous residence locations, immigration status, and a complete account of
traveling during the previous 3 years. The use of specific questionnaires aims at
excluding three types of infected donors:
1. Nonimmune travelers who acquired malaria abroad, preventing re-
lapses after specific treatment (P. vivax and P. ovale) or asymptomatic
parasitemia following inadequate chemoprophylaxis
2. Military personnel returning from endemic areas
3. Foreign visitors or immigrants from endemic regions who are usually
immune and asymptomatic
Although rules in different countries vary (16,25), blood donors are gener-
ally not accepted until 6 months after chemoprophylaxis has been finished in the
absence of symptoms. As a general policy, those with alleged past infection or
362 Wendel
who had taken chemoprophylaxis in the presence of symptoms are not accepted
during a 3-year period. This measure is highly effective for P. falciparum,
P. vivax, and P. ovale but not for P. malariae, for which longer periods of
infectivity are recorded. The questions apply only to products where viable red
cells are present, but may be disregarded for donations intended exclusively as
sources for plasma for fractionation. Since the world malarial zone is so vast, an
alphabetical list of countries reporting malaria transmission and a geographical
malarial map (provided by WHO) (4,5) should be present in every collection
facility. Unfortunately, avoiding infected donors only by using questionnaires
brings two additional problems (26):
1. Low specificityA study in the United States (12), where the incidence
of TTM is 0.25 cases/million units, claimed that reducing the interval
to 6 months after travel, irrespective of prophylaxis, would allow some
70,000315,000 (mean of 44,000) additional blood units to be collected
each year, with an additional calculated risk of 0.03 cases/million units
in the annual incidence. The authors stated that the 6-month deferral
was accurate for 88% of cases and the 3-year deferral period used for
preventing P. vivax and P. ovale (which accounted for only 33% of
cases) would have prevented only one case in 17 years.
2. Low sensitivityAfter a careful examination of all cases of TTM in the
United States, it was stated that at least 50% of them could be avoided
if the aforementioned questionnaire was correctly filled out (12);
additionally, the interviewer must rely upon the accuracy of the donors
history, which may be in error. This same study reported that a positive
history could be ascertained in only 2030% of the implicated donors,
since many of them were infected in childhood or do not remember
the episode.
In the light of these problems, some nonendemic countries have introduced
specific serological screening (see below), which is applicable only to selected
donors at increased risk, such as immigrants from or individuals born in endemic
areas or those who had malaria at least 3 years before donation (10,16,27). With
this strategy, about 10% of donors in France (10) who were tested by an indirect
immunofluorescence assay (IFAT) were confirmed as infective; in the United
Kingdom a recent work detecting antibodies by an antiglobulin ELISA (27) with
P. falciparum antigens reported a 98.5% saving of nonplasma components from
donors from tropical areas, which would lead to approximately 40,000 units
recovered annually. Other, more radical positions, such as a permanent exclusion
of all persons born or having lived in endemic areas, have been proposed, but they
should be carefully analyzed, especially in locations with blood shortage (28).
In countries where malaria is endemic, the exclusion for 3 years of donors
previously infected may be inappropriate. The use of some screening tests, such
Protozoan Parasites 363
Table 2 Proposed Action as the Result of a Positive Malaria ELISA Screening Test
for Donors Donating in Nonendemic Countries
G. Conclusion
A great deal of concern still exists related to the increase in incidence of TTM
for several reasons (3): increased travel by asymptomatic individuals from en-
demic areas to nonendemic regions (e.g., immune donors to a group of nonim-
mune recipients); the atypical incubation period (6 to >60 days); the lack of
awareness of attending physicians in most nonendemic countries, who may
additionally be unable to reach an accurate diagnosis when inadequate malarial
chemoprophylaxis is performed (which may change TTM natural evolution); the
ever-increasing appearance of drug-resistant strains, particularly of P. falciparum,
which causes the most severe cases; the resurgence of malaria in previously
eradicated areas; and, finally, the settlement of individuals in previously un-
habitated and virgin areas, mainly in South America, with a dramatic increase of
vector-transmitted malaria.
B. The Agent
Trypanosoma cruzi is a long and slender protozoa, belonging to the order
Kinetoplastida, family Trypanosomatidae, with a single nucleus, a flagellum, and
a kinetoplast, a DNA particle present in the mitochondria. There are three stages
in its evolutive life cycle.
A
Figure 3 (A and B) Trypomastigote forms present in the bloodstream from a patient with
acute transfusion-transmitted Chagas disease. This evolutive form is seen only in the acute
phase, where high parasitemia is usually present. In the chronic phase, this form is seldom
observed, usually recovered only by enrichment methods.
C. The Vector
The invertebrates responsible for transmission of T. cruzi are hematophagous bugs
belonging to the family Reduviidae and subfamily Triatominae (Fig. 4), with over
110 different species listed worldwide. Only 40 are adapted to human habitats,
and they are a significant vector for T. cruzi only in the New World, where they
can be detected from latitude 42N (northern California, Utah, Maryland) to
latitude 46S (Patagonia). They are known in English as the kissing bug or
cone-nose bug, in Spanish as vinchuca, and in Portuguese as barbeiro. The most
important reduviids are shown in the Table 3.
Figure 4 Chagas disease patterns of transmission. Sylvatic reservoirs (monkeys, marmosets, sloths,
armadillos, skunks) perpetuate the zonosis in the wild, while cats, dogs, rodents, and humans are the
main domestic reservoirs. In this setting some species have adapted to some very rudimentary huts,
where they are prone to feed on humans. If the blood meal derived from an already infected individual,
the infective cycle will be closed; additionally, infected persons may transmit Chagas disease through
blood transfusion, organ transplants, congenitally, breast feeding, or by accidental laboratory exposure,
Wendel
leading to new acute cases. Further details about transmission and life cycle can be found in the text.
Protozoan Parasites 371
Species Country
main reservoirs), some species are well adapted to very rudimentary human
dwellings made with wattle or bamboo, having unplastered walls with palm-
thatched roofs. These huts or shacks are usually found in isolated poor areas of
Latin America, grouped into small clusters or scattered around tiny villages.
After having a blood meal, the infected bug defecates in the sucking area,
leaving infective metacyclic trypomastigotes in feces, entering the host through
the feeding puncture or through mucosal membranes or by local scratching.
In humans, invasion into the macrophages occurs either via specific cellular
receptor or by endocytosis, and after a 20- to 30-hour period, a binary division
begins in the cytoplasm, generating 50500 intracellular amastigotes every 12
hours, with later differentiation to epimastigotes and trypomastigotes, which lyse
the infected cells, leading to release into the bloodstream. This, in turn, enables
the invasion of other cells, leading to host death or, more frequently, to the
development of an immune process that controls parasitemia, although with no
parasite eradication. Thus, parasitemia is present only in the acute process, while
in the chronic stage of Chagas disease, subpatent parasitemia is observed,
evidenced only by parasite-enrichment methods. There are no spontaneous cures
or normal relapses, except when a host immunosuppressive status occurs, such as
after organ transplants, oncological treatments, or in AIDS (73).
Chagas disease is also transmitted congenitally and by breast-feeding,
accidental laboratory contamination, organ transplants (74,75), or blood transfu-
sion (Fig. 4) (67).
372 Wendel
Figure 5 Estimated prevalence of Chagas disease among blood donors from 16 different
countries from the American continent, based on data from Table 4, and considering each
country as a whole, although marked regional differences are observed within each country
(Adapted from Refs. 63, 67, 80, 104.) Countries depicted in white have unknown data.
nents (up to 24 hours), although several patients with hemophilia treated only
with cryoprecipitate have been infected (94,95). In many places, blood transfu-
sion is recognized as the second most important way of transmission of Chagas
disease; on the other hand, it can be recognized as the main important route in
industrialized countries (e.g., Canada, the United States, and Spain) (5558,96).
The true number of reported cases is grossly underestimated, since no more
than 300 cases have been recently published in the literature (63,67), clearly
representing the tip of the iceberg. One reason for this is the lack of knowledge
or awareness of the disease (especially observed in industrialized, nonedemic
areas). In addition, there has been a decrease in observed cases in most developed
Protozoan Parasites 375
P = 1 (1f)n
where f = the prevalence of infected donors in the population and n = the number
of transfused units. However, no attention was paid to the probability of a patient
becoming infected when receiving one unit of infected blood; this is actually not
100%, but rather an average of 1225% although higher rates (47.6%) were
observed in Bolivia, a hyperendemic region (107). Thus, it seems rather prudent
to add to the original Cerisola formula the k factor for infectivity. It is not known
why the infectivity is not very high, but the low parasitemia rate (1 parasite per
20 mL of blood) and the simultaneous presence of inhibitory antibodies in the
plasma may be responsible. On the other hand, the survival rate (SR) of recipients
1 or 2 years after the transfusion event, averages 4050% (108). Therefore, the
376
Figure 6 The main pattern of human migration from Latin America to North America, Europe, Australia, and Japan, with the
corresponding expected legal immigrants in each region, although the actual number is expected to be more than this. (Based on Refs.
Wendel
63,67,77.)
Protozoan Parasites 377
P = 1 (1f)n k SR
In addition, one can speculate that the risk is also proportional to the ratio of
components produced (CP) from a single whole blood unit related to the number
of individual patients transfused with such units (PtTx), i.e., one whole blood unit
is transformed into x components that are transfused into y recipients.
Naturally, this additional index (CP/PtTx) should be included in the formula
above; however, it seems to be quite difficult to really estimate it, especially in
Latin America.
Adams-Stokes syndrome (due to right bundle branch block) may occur. Apical
aneurysm of the left ventricle with attached thrombi to the endocardium is often
found, leading to peripheral embolism.
The gastrointestinal system is compromised in 810% of patients, with
denervation of autonomic parasympathetic ganglia (Auerbachs plexus) with
esophageal or colonic hypotonia. Enlargement of internal organs (esophagus or
colon) are known as megasyndromes (e.g., megacolon or megaesophagus).
J. Preventive Measures
Prevention of Chagas disease can be accomplished by three different strate-
gies (119).
2. Serological Tests
Several method are available for screening, some of which are licensed in the
United States. Complement fixation was gradually replaced by IHA, IFA, or
ELISA, but none can be considered 100% sensitive. The possibility that serolog-
ical markers currently in use in blood banks (e.g., HBsAg, anti-HCV, -HIV,
-HTLV I/II, -HBc, ALT, or syphilis) could be used as surrogate markers for
T. cruzi was evaluated in 26,365 Brazilian donors (121). A very slight association
was observed only between syphilis in female donors (p = 0.005), but the low
number of cases (n = 4) precluded considering this to be a potential surrogate
once the same effect was not found in male donors or when the whole group was
considered. Thus, it seems that if T. cruziinfected donors are to be screened,
specific serological tests must be performed. Unfortunately, mandatory serologi-
cal screening is not a current approach in all Latin American countries, except for
Argentina, Brazil, Honduras, Paraguay, Uruguay, and Venezuela. Chile only
applies it in endemic regions. In Ecuador, although not mandatory, the majority
of Blood Services utilize testing.
3. Chemoprophylaxis
Because some countries still have a high prevalence of T. cruziinfected donors,
it is almost impossible to find enough noninfected donors to meet blood supply
needs. In addition, some of these areas are poor and devoid of a sophisticated
blood service. Also, some countries with a very low prevalence of infected donors
still do not consider Chagas disease a transfusion problem. Thus, it seems quite
reasonable to develop the sterilization of blood components, a procedure currently
under consideration in Bolivia, as advised by WHO (123). The first attempt to
promote sterilization of whole blood was by addition of thimerosal in 1952 (124).
Subsequently, other drugs were tested. An ideal model for screening drugs has
been developed, and more than 1000 drugs have been tested so far; among them,
only a few showed some trypanocidal effect. To have some value as a chemopro-
phylactic agent, a drug must fulfill the following criteria, irrespective of its mode
of action (125).
1. The drug must be active at pH 7.4 and at temperatures ranges from 30
to 22C, at which blood components are stored.
Protozoan Parasites 381
2. The drug must be active in undiluted blood and not adversely affect the
main metabolic and energetic pathways of red blood cells, platelets,
and plasma proteins throughout their storage period. In addition, no
interference must arise with the original antigenic composition of
human cells, nor should new antigens be generated.
3. The drug must not only be safe, but must also be used at a concentration
that, when transfused into the recipient, should not cause any pharma-
cological response for which it was originally designed.
Figure 7 Mechanism of action from crystal violet against Trypanosoma cruzi. CAT, Catalase; AH,
ascorbate; GSH, reduced gluthatione; SOD, superoxide dismutase. (Adapted from Refs. 126129.)
donors belonging to the chronic phase. It has been proved to be effective against
all parasite stages (amastigotes, epimastigotes, and trypomastigotes) (126,132)
and several different strains (e.g., Y, FL, G, J, M, Peru, and Sonya). In an
experimental study with 18 recipients transfused with seropositive treated blood,
none developed infection (133). Rezende et al. (134) reported that among a group
of 774 recipients transfused with unscreened blood components, at least 300 of
which could be assumed to derive from infected donors, none developed acute or
chronic infection. The Brazilian experience using over 50,000 units has confirmed
the efficacy of crystal violet treatment (129).
Side effects. There are some mild effects on red blood cells (Rouleaux
formation), but no changes in hemoglobin, pO2, pCO2, pH, Na+, or K+ have been
observed. ATP and 2,3 DPG levels are slightly decreased, but the results are not
statistically different from controls. The major imbalance of crystal violet occurs
Protozoan Parasites 383
K. Laboratory Screening
Several laboratory methods for blood donor screening are available.
1. Detection of Antibodies
Because of the easy application of the procedure and its relative low cost and high
sensitivity (though with variable specificity), methods aimed at T. cruzi antibody
detection are currently in use as a routine clinical diagnosis or as a blood donor
screening procedure. Several methods are commercially available in Latin Amer-
ica and the United States. The majority use whole parasites or crude lysates as
384 Wendel
the antigen source, although some research has been based on the development
of synthetic peptides or recombinant antigens. Trypanosoma cruzi antibody titer
is quite variable in an infected population and usually bears no relationship to
clinical symptoms; it also varies in the same individual during his or her lifetime.
The current serological methods are prone to cross-reacting with sera
containing antibodies against other infectious agents, such as yeasts, Leishmania
(Kala-azar and mucocutaneous leishmaniasis), T. rangeli (a nonpathogenic
agent found in Venezuela, Colombia, Peru, Ecuador, Panama, and Costa Rica),
T. gambiense (the agent of the sleeping sickness or African trypanosomiasis),
H. muscarum, L. seymoury, C. fasciculata, and other trypanosomatids. Addition-
ally, nonspecific IgM directed against phosphocoline (145), an antigenic fraction
widely present in several mycobacteria and parasites, is also responsible for
cross-reactivity, rendering the serological methods for T. cruzi antibody detection
still open for improvement. One has also to bear in mind that inconclusive results
are seen in approximately 35% of all blood donors, characterized by a reactive
result in only one assay (usually with clear negative or very low titer in a second
test). Even when a sample displays a clear reactive pattern, one cannot expect a
high positive predictive value. These events usually pose a major problem in
counseling of donors. For this reason, supplemental assays are also necessary
in order to confirm the screening tests. Finally, it is currently rather difficult
to obtain seroconversion panels as a result of the successful eradication of
disease in several countries, which makes the standardization of new tests a very
difficult task.
The main serological methods used for antibody detection are described in
the following sections (114,146).
agglutinins. Because of its relative high cost and the difficulty in finding suitable
commercially available reagents, this method is not used currently as blood
donor screening.
Indirect Hemagglutination Assay. Aqueous, soluble epimastigote extracts
can be easily fixed onto avian, sheep, or human red cells treated with tannic acid,
glutaraldehyde, or formaldehyde (147), maintaining satisfactory stability for
several months; several commercial kits are available. Although widely used
with easy performance, this method shows a lower sensitivity than IFAT or
ELISA (148150).
Indirect Immunofluorescence Assay. This test is based on fixed cultured
cells on slides and read by fluorescence microscopy (151,152). This very sensitive
test is usually the first to detect seroconversion samples (both IgM and IgG).
The practical performance of the test, its commercial availability, the long storage
period, relatively low cost, and high sensitivity were responsible for its wide use
as a screening test in Latin American countries since the mid-1970s. There are
two main problems with IFAT: the first is its nonapplicability to high-volume
screening. The second is its relatively high rate of false-positive results, since the
antibodies react mainly against intact membrane antigens, which are common to
other agents (especially Leishmania spp.) or epitopes displaying phosphocholine
(145). There is a wide array of autoimmune antibodies that also show consider-
able cross-reactivity (153).
ELISA. Since the early report by an immunoenzymatic test using crude
lysates (154) with some problems in its early phase (155,156) several important
improvements have been developed, including more purified antigens that en-
hanced the sensitivity and specificity of the test (157159). Recombinant proteins
or synthetic peptides show better specific results than parasite extracts (lower
cross-reactions) (160,161) but still have variable sensitivities according to the
patient and clinical manifestation of the disease, but still have to enhance the
sensitivity, an alternative would be to combine several recombinant antigens or
synthetic peptides to sensitize microwells. However, some interferences, such as
histeric hindrance, leads to poorer results for the mixed antigens than for
individual antigens (159,162), still leaving this alternative for the future. The pos-
sibility of automation and large use to several samples make this method as the
most reliable one for screening blood donors. Several kits are commercially
available, including in the United States, all based in epimastigote antigens.
2. Supplemental Tests
As previously discussed, there is still a need for good supplemental assays in
order to validate the original antibody screening tests used in the blood bank.
Currently, two assays are under evaluation.
386 Wendel
Western Blot and Line Immunoassay. Crude or purified antigens are run
in polyacrilamide gel electrophoresis and then transferred onto nitrocellulose
strips. All reactive bands are detected by an immunoenzymatic reaction. Although
this method is widely used for viral tests, there are still several problems when it
is applied to T. cruzi. The first one is the large number of bands present in the test,
according to each antigenic preparation, with some authors reporting up to 15 or
20 different bands ranging from 10 to 300 kDa; in addition, two-dimensional
electrophoresis reveals the presence of different proteins with the same molecular
weight (163). The second problem is that several antibodies are not quite specific
and when eluted from a single band and put again to react against a new
nitrocellulose strip, specimens will react against bands other than those originally
eluted (164). The sensitivity of Western blot has been reported to range from 86.8
to 100% (116,165,166). Highly purified antigens (natural or recombinant) or
synthetic peptides on nitrocellulose strips have been developed either in combi-
nation or individually, as single (167) or multiple line assays (168170).
Radioimmunoprecipitation Assay. Cultured epimastigotes, when placed
in a medium containing a radioisotope (125I or 35S), will incorporate radioactivity,
which will be demonstrated when lysed forms are reacted against specific
antibodies present in the serum. Two major glycoprotein bands are related to a
positive result: gp 72 and gp 90 (171). With some slight modifications in the
principle, there are three groups in the United States (87, 171,172) currently in
the process of validating the method; however, due to its complexity and high
cost, it will be restricted to only a few labs.
PCR. Chagas disease is a model of intense PCR efforts due to the relative
low sensitivity and technical difficulties involved with xenodiagnosis of and
hemoculture attained from chronic patients; in addition, both methods require as
long as 120 days to reach a conclusive result.
The main difficulty faced in applying PCR for the detection of Chagas
disease in chronic patients derives from the low and intermittent parasitemia
observed in an important fraction of chronic carriers. In order to obviate that,
investigators have focused on DNA sequences largely represented on the T. cruzi
genome, such as on the kinetoplast (141). Even though no methodology has
achieved 100% sensitivity compared to serology of chronic patients, the results
of all published comparative studies clearly favor the use of PCR for confirmation
of a serology-positive result for Chagas disease.
One problem still unsolved is the large blood volume that must be sampled
and extracted to detect T. cruzi DNA by PCR. PCR methods achieve a sensitivity
of about one parasite in 20 mL of blood, but chronic carriers may harbor a
parasitemia less than that amount.
There has been a report of a PCR method based on DNA extracted from
serum that achieved similar results when whole blood DNA was obtained
Protozoan Parasites 387
from the same samples, which led investigators to suggest that serum could
replace whole blood for PCR detection of T. cruzi (142). These results must be
interpreted with caution based on the fact that parasites tend to remain on the
buffy coat of separated blood, as observed by many groups.
PCR is currently applied for the diagnosis of Chagas disease in both
chronic and acutely infected patients and is under scrutiny as a confirmatory test
for blood banks. It is also the method of choice for testing chemotherapeutic
agents for T. cruzi on experimentally infected animals (143), and a quantitative
PCR method has been developed that can be used to monitor patients during
treatment (144).
L. Therapy
Treatment of acute cases is done by controlling the symptoms and by the use of
nifurtimox or benznidazole (173). These drugs are effective only in the acute
phase and should be used immediately after diagnosis; benznidazole might be
effective in the chronic phase in infected children (174), though it is not devoid
of severe side effects. The clinical management of chronic Chagas disease is
directed toward the control of signs and symptoms, but no permanent cure can
be achieved.
M. Conclusion
As in malaria, several problems related to the geographical distribution, pattern
of emigration, high number of asymptomatic donors, lack of medical knowledge
to correctly diagnose most of the cases, particularly in developed countries, and
the still pending technical refinements for laboratory screening result in Chagas
disease remaining a major public health problem in all American countries.
In addition, even with complete vectorial elimination, Chagas disease will re-
main as a zoonosis. Millions are already infected, requiring medical attention
throughout their lifetime. Finally, some asymptomatic patients are women of
childbearing age, who will continue to propagate the disease congenitally. Never-
theless, continuous efforts in several countries are yielding positive results;
complete elimination of the main vector is expected in fewer than 10 years. Also,
one can speculate that, given the association between exposure and age (120,121),
it is likely that no further vectorially contaminated donors younger than 30 years
of age will be found within 1020 years in Brazil (175). Thus, it seems that the
strategies currently developed to prevent transfusional transmission of Chagas
disease will certainly need a reassessment in a decade. This is a remarkable feat,
because only 100 years will have elapsed between the discovery and elimination
of Chagas disease, an unprecedented accomplishment in the history of human
infectious diseases.
388 Wendel
REFERENCES
1. Woolsey G. Transfusion for pernicious anaemia: two cases. Ann Surg 1911;
53:132135.
2. Turc JM. Malaria and blood transfusion. In: Westphal RG, Carlson KB, Turc JM,
eds. Emerging Global Patterns in Transfusion-Transmitted Infections. Arlington,
VA: AABB, 1990:3143.
3. International Forum: Which are the appropriate modifications of existing regulations
designed to prevent the transmission of malaria by blood transfusion, in view of the
increasing frequency of travel to endemic areas? Vox Sang 1987; 52:138148.
4. WHO. World malaria situation in 1990. Weekly Ep Rec 1992; 67:161168.
5. WHO. World malaria situation in 1990. Weekly Ep Rec 1992; 67:169176.
6. Gilles HM, Warrell DA. Bruce-Chwatts Essential Malariology. 3rd ed. London:
Edward Arnold, 1993.
7. Bruce-Chwatt LJ. Transfusion malaria revisited. Trop Dis Bull 1982; 79:827840.
8. Bruce-Chwatt LJ. Transfusion malaria. Bull WHO 1974; 50:337346.
9. Bruce-Chwatt LJ. Blood transfusion and tropical disease. Trop Dis Bull 1972;
69:825862.
10. Chataing B. Prevention du paludisme transfusionel. Rev Franc Transf Immuno-
hematol 1988; 31:8188.
11. Tabor E. Infectious Complications of Blood Transfusion. New York: Academic
Press, 1982.
12. Nahlen BL, Lobel HO, Cannon SE, Campbell CC. Reassessment of blood donor
selection criteria for the United States travellers to malarious areas. Transfusion
1991; 31:798804.
13. WHO. The biology of malaria parasites. Technical Report Series 743. Geneva:
World Health Organization, 1987.
14. Lpez-Antuano FJ, Schmunis G, eds. Diagnosis of malaria. Scientific Publication
No. 512. Washington, DC: Pan American Health Organization, 1990.
15. Fairley NH. Trans R Soc Trop Med Hyg 1947; 40:621676.
16. Council of Europe. Guide to the preparation, use and quality assurance of blood
components. Strasbourg, 1992: 17.
17. Wells L, Ala FA. Malaria and blood transfusion. Lancet 1985; i:13171318.
18. Andrade JCR, Wanderley DMV. Malria induzida no Estado de So Paulo, Brasil.
Rev Soc Bras Med Trop 1991; 24:157161.
19. Ferreira MV, Camargo LMA, Carvalho ME, Ninomia RT, Garcia LAV, Santos FR.
Prevalence and levels of IgG and IgM antibodies against Plasmodium falciparum
and P. vivax in blood donors from Rondonia, Brazilian Amazon. Mem Inst Oswaldo
Cruz 1993; 88(2):263269.
20. Kiesslich D, Torres KL, Fraiji NA. Mtodos de triagem para Malria em Doadores
de Sangue de rea Endmica. Bol Soc Bras hematol Hemot 1997; 19:2127.
21. Salutari P, Sica S, Chiusolo P, Micciulli G, Plaisant P, Nacci A, Antinori A, Leone
G. Plasmodium vivax malaria after autologous bone marrow transplantation: an
unusual complication. Bone M Transpl 1996; 18:805806.
22. Talabiska DG, Komar MJ, Wytock DH, Rubin RA. Post-transfusion acquired
Protozoan Parasites 389
in well defined population groups in an endemic area of Brazil. Rev Inst Med Trop
So Paulo 1994; 36:157162.
41. Hong VT, VanBe T, Tran PN, Thanh LT, Hien LV, OBrien E, Morris GE. Screening
donor blood for malaria by polymerase chain reaction. Trans R Soc Trop Med Hyg
1995; 89:4447.
42. Chikwem JO, Mohammed I, Okara GC, Ukwandu NCD, Ola TO. Prevalence of
transmissible blood infections among blood donors at the University of Maiducuri
teaching hospital, Maiduguri, Nigeria. East Afr Med J 1997; 74:213216.
43. Chagas C. Nova espcie morbida do homem, produczida por um Trypanosoma
(trypanosoma cruzi): Nota prvia. Brasil Md 1909; 23:161.
44. Chagas C. Neue Trypanosomen. Arch Schiffs Tropenhyg 1909; 13:120122.
45. Chagas C. ber eine neue Trypanosomiasis des Menschen. Arch Schiffs Tropenhyg
1909; 13:351353.
46. Chagas C. Nova tripanozomiase humana. Estudos sobre a morfolojia e o ciclo
evolutivo do Schizotrypanum cruzi n.g., n.s.p., ajente etioljico de nova entidade
mrbida no homem. Mem Inst Oswaldo Cruz 1909; 1:159218.
47. Chagas C. Nova entidade mrbida do homem. Resumo geral de estudos etioljicos
e clnicos. Mem Inst Oswaldo Cruz 1911; 3:219275.
48. Chagas C. The discovery of Trypanosoma cruzi and of American trypanosomiasis.
Mem Inst Oswaldo Cruz 1922; 15:111.
49. Chagas Filho C. Carlos ChagasOfcina Grfica da Universidade do Brazil, 1959.
50. Mazza S, Montana A, Benitez C, Juzin E. Transmisin de Schizotrypanum cruzi
al nio por leche de la madre con enfermedad de Chagas. Publ MEPRA 28:4146.
51. Pellegrino J. Transmisso da doena de Chagas pela transfuso de sangue. Primeiras
comprovaes sorolgicas em doadores e candidatos a doadores de sangue. Rev
Bras Med 1949; 6:297301.
52. Faria P. Sifilis, Maleita, Doena de Chagas e Transfuso. Folia Clin Biol 1951;
17:113117.
53. Freitas, JLP, Amato V, Sonntag R, Biancalana A, Nussenszweig V, Barreto JG.
Primeiras verfcaces de transmisso acidental da molstia de Chagas ao homem
por transfuso de sangue. Rev Paul Med 1952; 40:3640.
54. Nussenzweig V, Sonntag R, Biancalana A, Freitas JLP, Amato Neto V, et al. Ao
de corantes tri-fenil-metanicos sobre o Trypanosoma cruzi in vitro. Emprego da
Violeta de Genciana na proflaxia da transmisso da Molstia de Chagas por
transfuso de sangue. O Hospital 1953; 44:731744.
55. Geiseler PJ, Ito JI, Tegtemeier BR, Kerndt PR, Krance R. Fulminant Chagas disease
in bone marrow transplantation. 27th Intersc. Conf. on Antimicro Agents and
Chemoter. Washington: Am Soc for Microbiol 1987:418.
56. Grant I, Gold JWH, Wittnee M, et al. Transfusion associated acute Chagas disease
acquired in the United States. Ann Intern Med 1989; 111:849851.
57. Nickerson P, Orr P, Schoeder MC, Sekla L, Johnson J. Transfusion associated
Trypanosoma cruzi infection in a non endemic area. Ann Intern Med 1989;
111:851853.
58. Cimo PL, Luper WE, Scouros MA. Transfusion-associated Chagas disease in
Texas: report of a case. Tex Med 1993; 89:4850.
59. Lenes BA, Leiby DA, Tibbals MA, Olmedo M. A prospectively identified case of
Protozoan Parasites 391
75. Ferraz AS, Figueiredo JFC. Transmission of Chagas disease through transplanted
kidney: occurrence of the acute form of the disease in two recipients from the same
donor. Rev Inst Med Trop S Paulo 1993; 35:461463.
76. Dias JCP, Brener Z. Chagas disease and blood transfusion. Mem Inst Oswaldo Cruz
1984; 79:139147.
77. Schmuis GA. Trypanosoma cruzi, the etiologic agent of Chagas disease: status in
the blood supply in endemic and non endemic countries. Transfusion 1991; 31:547
557.
78. Wendel S, Gonzaga AL. Chagas disease and blood transfusion: a new world
problem? Vox Sang 1993; 64:112.
79. Schmunis GA. Tripanosomase Americana: seu impacto nas Amricas e perspectivas
de eliminaco. In: Dias JCP, Coura JR, eds. Clinica e teraputica da Doena de
Chagas. Rio de Janeiro: Edotora Fiocruz, 1997:411427.
80. Wendel S. Doena de Chagas Transfusional. In: Dias JCP, Coura JR, eds. Clinica e
teraputica da Doena de Chagas. Rio de Janeiro: Edotora Fiocruz, 1997:411427.
81. Wendel S, Neitzert E, Lopes VHG, Batemarchi MV. Evaluation of different screen-
ing tests for T. cruzi antibodies and correlation with immunoblotting technique. Rev
Paul Med 1992; 110: Abstract TTD17.
82. Trujillo FC, Kasten FL, Gutirrez MMS, Gutirrez RH. Prevalencia de infeccin a
Trypanosoma cruzi en donadores de sangre en el estado de Jalisco, Mexico. Rev
Soc Bras Med Trop 1993; 26:8992.
83. Ache A. Prevalencia de infeccin humana por Trypanosoma cruzi en Bancos de
Sangre en Venezuela. Rev Inst Med Trop S Paulo 1993; 35:443448.
84. Kerndt PR, Waskin HA, Kirhhoff LV, Steurer F, Waterman SH, Nelson JM, Gellert
GA, Shulman IA. Prevalence of antibody to Trypanosoma cruzi among blood
donors in Los Angeles, California. Transfusion 1991; 31:814818.
85. Pan AA, Brashear J, Schur JD, Winkler MA, Hall H, Shih J, Decker R. Prevalence
study and confirmation of seropositive antibodies to Trypanosoma cruzi (Chagas
disease) in Hispanic and Non-Hispanic blood donors in the United States. Rev Paul
Med 1992; 110(5) (suppl.): Abstract TTD-18.
86. Read EJ, Leiby DA, Dodd RY. Seroprevalence of Trypanosoma cruzi (T. cruzi) in
blood donors with and without risk infection. Blood 1994; 84 (suppl 1): Abstract
1853.
87. Winkler MA, Brashear RJ, Hall HJ, Schur JD, Pan AA. Detection of antibodies
to Trypanosoma cruzi among blood donors in the southwestern and western
United States. IIEvaluation of a supplemental enzyme immunoassay and radio-
immuneprecipitation assay for confirmation of seroreactivity. Transfusion 1995;
35:219225.
88. Leiby DA, Read EJ, Lenes BA, Yund AJ, Stumpf RJ, Kirchhoff LV, Dodd RY.
Seroepidemiology of Trypanosoma cruzi, etiologic agent of Chagas disease, in US
blood donors. J Infect Dis 1997; 176:10471052.
89. Amato Neto V, Leonhardt H, Souza HBWT. Liofilizao do plasma: Medida capaz
de evitar a transmisso da Doena de Chagas em Bancos de Sangue. Rev Inst Med
trop So Paulo 1966; 8:122124.
90. Sullivan TS. Viability of Trypanosoma cruzi in citrated blood stored at room
temperature. J Parasitol 1944; 30:200.
91. Weinman D, MacAlister J. Prolonged storage of human pathogenic protozoa with
conservation of virulence. Am J Hyg 1947; 45:102121.
Protozoan Parasites 393
against Trypanosoma cruzi in the blood by ascorbate. Mol Biochem parasitol 1988;
27:241248.
129. Souza HM. The present state of chemoprophylaxis in transfusional Chagas disease
(editorial). Rev Soc Bras Med Trop 1989; 22:13.
130. Ramirez LE, Lages-Silva E, Painetti GM, Rabello RMC, Bordin JO, Souza HM.
Prevention of transfusion-associated Chagas disease by sterilization of Trypano-
soma cruzi-infected blood with gentian violet, ascorbic acid, and light. Transfusion
1995; 35:226230.
131. Amato Neto V, Mellone O. Estudo sobre a efccia da Violeta de Genciana na
proflaxia da transmisso da doena de Chagas em bancos de sangue: investigao
em voluntrio, receptor de sangue de caso agudo, ao qual foi adicionado o corante.
O Hospital 1959; 55:343346.
132. Schlemper BR Jr. Estudos experimentais de Quimio-proflaxia de transmisso da
doena de Chagas por transfuso sangnea. Rev Pat Trop 1978; 7:55111.
133. Amato Neto V. Contribuio ao conhecimento da forma aguda da Doena de
ChagasTese: Faculdade de Medicina da Universidade de So Paulo, 1958.
134. Rezende JM, Zupelli W, Bafutto M. O problema da transmisso da doena de
Chagas por transfuso de sangue. Emprego da Violeta de Genciana como medida
profltica. Ver Goiana Med 1965; 11:3547.
136. Docampo R, Moreno SNJ. The metabolism and mode of action of gentian violet.
Drug Met Rev 1990; 22:161178.
137. Gottlieb P, Shen LG, Chimezie E, Bahng S, Keeney ME, Horowitz B, Bem-Hur E.
Inactivation of Trypanosoma cruzi trypomastigote forms in blood component
by photodynamic treatment with phthalocyanines. Photochem Photobiol 1995;
62:869874.
138. Wendel S, Takaoka DT, Jancs MM, Mariano M, Fragata Filho AA, Pereira-
Chioccola Lourenco AM. A possible role of leukocyte filters to prevent transfusion-
transmitted Trypanosoma cruzi. Vox Sang 1994; 67(suppl 2):22.
139. Souza HM, Bordin JO, Bordossy L, MacPhereson DW, Blajchman MA. Prevention
of transfusion-associated Chagas disease: efficacy of white cell-reduction filters in
removing Trypanosoma cruzi from infected blood. Transfusion 1995; 35:723726.
140. Wendel S, Takaoka DT, Jancs MM, Mariano M, Leite KM, Chioccola VLP,
Loreno AM, Fragata Filho AA. Is there a role for leukocyte depletion filters to
remove Trypanosoma cruzi infection from donor blood? Bol Soc Bras Hematol
Hemot 1995; 17:2531.
141. Moser DR, Kirchhoff LV, Donelson JE. Detection of Trypanosoma cruzi by DNA
amplification using the polymerase chain reaction. J Clin Microbiol 1989; 27:1477
1482.
142. Russomando G, Figueredo A, Almirn M, Sakamoto M, Morita K. Polymerase
chain reaction-based detection of Trypanosoma cruzi DNA in serum. J Clin
Microbiol 1992; 30:28642868.
143. Urbina J, Payares G, et al. Cure of short and long-term experimental Chagas disease
using D0870. Science 1996; 273:969971.
144. Centurion-Lara A, Barrett L, van Voohis WC. Quantitation of parasitemia by
competitive polymerase chain reaction amplification of parasite kDNA minicircles
396 Wendel
during chronic infection with Trypanosoma cruzi. J Infect Dis 1994; 170:1334
1338.
145. Lal RB, Ottesen EA. Phosphocoline epitopes on helminth and protozoal parasites
and their presence in the circulation of infected human patients. Trans Roy Soc Trop
Med Hug 1989; 83:652655.
146. Ferreira AW. Serological diagnosistests for Chagas disease serodiagnosis: a
review. In: Wendel S, Brener Z, Camaro ME, Rassi A, eds. Chagas Disease
(American Trypanosomiasis): Its Impact on Transfusion and Clinical Medicine. So
Paulo: ISBT, 1992: 179193.
147. Sasaki AT, Hoshino-Shimizu S, Nakamura PM, Vaz AJ, Camargo ED, Silva MD.
Sorodiagnstico da doena de Chagas: novo reagente para o teste de Hema-
glutinao indireta (THAIIAL). Rev Soc Bras Med Trop 1996; 29:137144.
148. Luquetti AO. Use of Trypanosoma cruzi defined proteins for diagnosismulticentre
trial, serological and technical aspects. Mem Inst Oswaldo Cruz 1990; 85:497505.
149. Zicker F, Smith PG, Luquetti AO, Oliveira OS. Mass screening for Trypanosoma
cruzi infections using the immunofluorescence, ELISA and hemagglutination tests
on serum samples and on blood eluates from filter paper. Bull WHO 1990;
68:465471.
150. Andrade ALSS, Martelli CMT, Luquetti AO, Oliveira OS, Silva SA, Zicker F.
Serologic screening for Trypanosoma cruzi among blood donors in central Brazil.
Bull Pan Am Health Organ 1992; 26:157164.
151. Fife EH Jr, Mushel LH. Fluorescent antibody technique for serodiagnosis of
Trypanosoma cruzi infection. Proc Soc Exp Biol NY 1959; 101:540543.
152. Camargo ME, Souza SL. The use of filter paper blood smears in a practical
fluorescent test for American trypanosomiasis serodiagnosis. Rev Inst Med Trop S
Paulo 1966; 8:255258.
153. Velasquez L, Reyes L, Thors C, Miettinen A, Chinchella M, Linder E. Autoantibod-
ies give false positive reactions in the serodiagnosis of Trypanosoma cruzi. Trans
Roy Soc Trop Med Hyg 1993; 87:35.
154. Voller A, Draper C, Bidwell DE, Bartlett A. A micro-plate enzyme-linked im-
munosorbent assay (ELISA) for Chagas disease. Lancet 1975; 1:426429.
155. Wendel S, Lopes VHG, Batemarchi MV, Takaoka DT, Neitzert E. High levels of
false positive reaction for Trypanosoma cruzi antibodies tests used for blood donors
screening. Transfusion 1993; 33(suppl):41S.
156. Carvalho MR, Krieger MA, Almeida E, Oelemann W, Shikanai-Yasuda MA,
Ferreira AW, Pereira JB, Sez-Alquzar A, Dorlhiac-Llacer PE, Chamone DF,
Goldenberg S. Chagas disease diagnosis: evaluation of several tests in blood bank
screening. Transfusion 1993; 33;830834.
157. Ferreira AW, Belem ZR, Moura MEG, Camargo ME. Aspectos da padronizao de
testes sorolgicos para a Doena de Chagas: Um teste imunoenzimtico para a
triagem de doadores de sangue. Rev Inst Med Trop S Paulo 1991; 33:123128.
158. Brashear RJ, Winkler MA, Schur JD, Lee H, Burczak JD, Hall HJ, Pan AA.
Detection of antibodies to Trypanosoma cruzi among blood donors in the south-
western and western United States. I. Evaluation of the sensitivity and specificity of
an enzyme immunoassay for detecting antibodies to T. cruzi. Transfusion 1995;
35:213218.
Protozoan Parasites 397
159. Oelemann WMR, Teixeira MGM, Costa GCV et al. Evaluation of three commer-
cial enzyme-linked imunosorbent assays for diagnosis of Chagas disease. J Clin
Microbiol 1998; 36:24232427.
160. Houghton RL, Benson D, Skeiky Y, Sleath P, Lodes M, Badar R, Krettli A, Reed
SG. Multi-epitope peptide elisa for detection of serum antibodies to Trypanosoma
cruzi in patients with treated and untreated Chagas disease. Transfusion 1996;
36(suppl):137.
161. Hoffman A, Jaczko B, Jones J, Tursi N, Lee SR. Evaluation of a prototype screening
ELISA and supplemental analysis for the serological identification of Chagas
disease. Transfusion 1997; 37(suppl):181.
162. Umezawa ES, Bastos SF, Camargo ME, Yamauchi LM, Santos MR, Gonzales A,
Zingales B, Levin MJ, Souza O, Rangel-Ado R, Franco da Silveira J. Evaluation
of recombinant antigens for serodiagnosis of Chagas disease in South and Central
America. J Clin Microbiol 1999; 37:15541560.
163. Lanar DE, Manning JE. Major surface proteins and antigens on the different in vivo
and in vitro forms of Trypanosoma cruzi. Mol Biochem Paras 1984; 11:119131.
164. Rosfjord EC, Mikhael KS, Rowland EC, Powell MR. Analysis of antibody cross-
reactivity in experimental american trypanosomiasis. J Parasitol 1990; 76:698702.
165. Reiche EMV, Cavazzane M Jr., Okamura H, Tagata EC, Jankevicius SI, Jankevicius
JV. Evaluation of the Western blot in the confirmatory serologic diagnosis of
Chagas disease. Am J Trop Med Hyg 1998; 59:750756.
166. Teixeira MGM, Borges-Pereira J, Neitzert E, Souza MLNX, Peralta JM. Develop-
ment and evaluation of an enzyme linked immunotransfer blot technique for
serodiagnosis of Chagas disease. Trop Med Parasitolo 1994; 45:308312.
167. Pastini AC, Iglesias SR, Carricate VC, Guerin ME, Sanchez DO, Frasch AC.
Immunoassay with recombinant Trapanosoma cruzi antigens potentially useful
for screening donated blood and diagnosing Chagas disease. Clin Chem 1994;
40:18931897.
168. Sabino EC, Salles N, Stoops E, Santos ML, Saez-Alquezar A. Evaluation of Chagas
Innolia assay as a confirmatory test for Chagas disease. Mem Inst Oswaldo Cruz
1997; 92(suppl 1):266.
169. Costa GCV, Teixeira MGM, Borges-Pereira J, Castro JAF, Coura JR, Vanderborght
BOM, Stroops E, Zrein M, Peralta JM, Oelemann WMR. A recombinant antigen
and peptide line immunoassay (LIA) as an alternative diagnostic test for Chagas
disease. Mem Inst Oswaldo Cruz 1997; 92(suppl 2):267.
170. Oelemann WMR, Vanderborght BOM, Verssimo da Costa GC, Teixeira MGM,
Borges-Pereira J, Castro JAF, Coura JR, Stoops E, Hulstaert F, Zrein M, Peralta JM.
A recombinant peptide antigen line immunoassay optimized for the confirmation of
Chagas disease. Transfusion 1999; 39:711717.
171. Kirchhoff LV, Gam AA, Gusmo RA, Goldsmith RS, Rezende JM, Rassi A.
Increased specificity of serodiagnosis of Chagas disease by detection of antibody
to the 72- and 90-kilodalton glycoproteins of Trypanosoma cruzi. J Infect Dis 1987;
155:561564.
172. Leiby DA, Wendel S, Takaoka DT, Fachini R, Oliveira LC, Tibbals MA. Confirma-
tory testing for Trypanosoma cruzi antibodies: validation of radioimmuneprecipita-
tion (RIPA) testing. Transfusion 1997; 37(suppl 95):5301.
398 Wendel
173. Rassi A, Luquetti AO. Therapy of Chagas disease. In: Wendel S, Breuer Z, Camargo
ME, Rassi A, eds. Chagas Disease (American Trypanosomiasis): Its Impact on
Transfusion and Clinical Medicine. So Paulo: ISBT, 1992:237247.
174. Andrade ALLS, Zicker F, Oliveira RM, Silva AS, Luquetti AO, Travassos LR,
Almeida IC, Andrad SS, Andrade JG, Martelli CMT. Randomised trial of efficacy
of benznidazole in treatment of early Trypanosoma cruzi infection. Lancet 1996;
348:14071412.
175. Dias JCP, Schofield CJ. Controle da transmisso transfusional da doena de Chagas
na Iniciativa do Cone Sul. Rev Soc Bras Med Trop 1998; 31:373383.
17
Tickborne Infections
Ritchard G. Cable
American Red Cross Blood ServicesConnecticut Region,
Farmington, Connecticut
Jonathan Trouern-Trend
Epidemiology and Surveillance Program, American Red Cross
ARCNET, Farmington, Connecticut
I. INTRODUCTION
399
400 Cable and Trouern-Trend
these organisms efficient vectors of disease. First, the ingestion of host blood
allows collection of pathogens in the body of the tick. If the tick subsequently
moves to another host, transmission may occur. The long period of attachment on
a host increases the likelihood of transmission. High reproductive potential
ensures high populations of ticks and increases possibility of disease transmis-
sion. Pathogens are transmitted between life stages in some ticks (e.g., between
the larva and the nymph), a phenomenon known as transstadial transmission.
Pathogens may also be passed to the next generation by transovarial transmission.
Both types of transmission are important in the maintenance of pathogens in tick
populations (2).
Mites belong to the same order as ticks and share many of the same
characteristics and parasitic lifestyles. Ticks are really giants in the mite family,
most mites being much smaller than the smallest ticks (5). Scrub typhus, which
is miteborne, is discussed here since it may be transfusion-transmitted.
A great many tickborne diseases have been described. A number of these
have the potential for transfusion transmission. The more important tickborne
diseases of interest in transfusion medicine are summarized in Table 1. The re-
mainder of the chapter will expand upon these diseases.
A. Flaviviruses
These enveloped single-stranded RNA viruses replicate in the cytoplasm. The
Flavivirus family contains nonarborviruses such as hepatitis C and hepatitis G
(7,8). Only one of the tickborne flaviviruses, the central European subtype of
tickborne encephalitis, has been documented as causing transfusion-transmitted
disease (9). Theoretically, others could do so. Two cases of tickborne viral en-
402 Cable and Trouern-Trend
Transfusion-
Agent Disease transmitted? Country Ref.
Viruses
Tickborne encepha- Kumlinge disease, Yes Finland 8,9
litis virus tickborne
encephalitis
Deer tick virus None known Possible United States 11
Colorado tick fever Colorado tick fever Yes United States 12,13
virus
Bacteria
Borrelia burgdorferii Lyme disease Possible United States 1719
Borrelia recurrentis Relapsing fever Possible Africa, China 2427
Rickettsia
Rickettsia rickettsi Rocky Mountain Yes United States 28
spotted fever
Orienta tsutsugamushi Scrub typhus Possible Asia, 30
Australia
Ehrlichia chaffiensis Human monocytic Possible United States 31
ehrlichiosis
Ehrlichia spp. Human granulo- Possible United States 31,32
cytic ehrlichiosis
Protozoa
Babesia microti Babesiosis Yes United States 3336,
6368
WA1 Babesiosis-like Yes United States 37,38
MO1 Babesiosis-like Possible United States 39
TW1 None known Possible Taiwan 40
cephalitis (Kumlinge disease) were reported from a single donor from Kumlinge,
Finland. The disease, endemic on this isolated island, was reportedly transmitted
by a blood donation a few hours before the donor became ill. The transfusion
illness was indistinguishable from the tickborne illness. No details on the blood
components storage or transfusion were published, and it is not clear how the
disease was recognized. The recipients recovered.
Another flavivirus, deer tick virus, was described in 1997. It has been
found in Ixodes scapularis, a vector of several human diseases, including Lyme
disease and babesiosis (10,11). This virus has not been demonstrated to cause
disease in humans. However, its close relationship to other viruses in the
tickborne encephalitis complex warrants some concern. In a serosurvey of
ticks from New England, a small percentage was found to be harboring this virus
Tickborne Infections 403
(11). Further studies will be needed to determine whether this virus is a public
health problem.
B. Retroviruses
This family contains enveloped double-stranded RNA viruses that replicate in the
cytoplasm. Ticks spread several of these viruses to humans. Most are responsible
for only a few cases of disease, although Colorado tick fever virus causes several
hundred cases a year in North America.
Only Colorado tick fever virus has been demonstrated to cause transfusion-
transmitted disease. Cases of Colorado tick fever occur in the Rocky Mountains
and Pacific slope of the western United States and Canada. The vector is the wood
tick, Dermacentor andersoni. Up to 15% of humans at high risk (forest rangers,
loggers, back-country hikers) have antibodies to the virus. Most cases occur
between April and July, mirroring tick activity. Several hundred cases are reported
each year. Since reporting requirements vary by region, cases are thought to be
significantly underestimated. Cases have been imported to other parts of the
United States by people traveling in the endemic areas. Incubation is usually 35
days after a tick bite, sometimes up to 14 days. No prodromal manifestations
occur. Onset of symptoms is rapid. Fever, headache, and myalgia are the most
common symptoms. Encephalitis, thrombocytopenia, disseminated intravascular
coagulation (DIC), and a hemorrhagic state resembling Dengue hemorrhagic
fever have occurred. Many individuals, especially adults over 30 years, experi-
ence a prolonged convalescence (12).
Colorado tick fever was transmitted in Montana from a healthy blood donor
who developed fever 18 hours after blood donation and 4 days after removing a
tick (13). The blood was transfused during surgery to an 82-year-old with bowel
obstruction, who developed a prolonged febrile illness lasting 23 days. Both
donor and recipient blood were demonstrated to have Colorado tick fever virus
by mouse inoculation, confirming the diagnosis. This case illustrates again the
importance of reporting postdonation illness in order to recognize these rare
disease transmissions.
The virus can be isolated from plasma for up to 7 days after the onset of
symptoms and up to 20 weeks in the red cell fraction. An infection usually confers
immunity; however, rare cases of reinfection have been reported.
in the United States and western Europe. This disease accounts for more than 90%
of arthropodborne diseases reported in North America each year. In the United
States, endemic foci exist along the Atlantic seaboard from Massachusetts to
Maryland, in the upper Midwest (Wisconsin and Minnesota), and in the West
(California and Oregon). Cases have been reported from 45 states as well as the
Canadian provinces of British Columbia and Ontario (14). Elsewhere, cases have
been seen in the former Soviet Union, China, Japan, and Australia. The main
vectors are four host Ixodid ticks: I. scapularis (also called I. dammini) in the
Eastern and midwestern United States, I. pacificus in the western United States,
I. ricinus in Europe, and I. persulcatus in Asia.
In North America, the spirochete responsible for Lyme disease circulates
between the white-footed mouse (Peromyscus leucopus) and the white-tailed deer
(Odocoileus virginianus) via I. scapularis. The transmission cycle of the spiro-
chete starts with an infected mouse. Larval ticks hatch in the spring or early
summer and search out their first host, usually a mouse. After feeding for about
2 days, the tick drops off. The tick does not feed again until the next spring, after
it has molted to the nymphal stage. Usually the nymph feeds once, again usually
on a mouse, then molts to an adult. If the preferred host cannot be found, the
nymph will feed on other mammals, including humans. Nymphal ticks, infected
from their first blood meal, transmit the spirochete to humans. The spirochete
enters the hosts body at the site of the tick bite, then travels via the blood and
lymphatics to other organs of the body, where pathogenesis occurs. Adult ticks
feed on deer, then lay eggs. This association between Lyme disease and deer is
responsible for the dramatic increase in cases in this century. The abundance of
the vector correlates with the number of deer in the area.
Lyme disease can be divided into three stages. In the first stage there is a
localized skin lesion at the bite site called erythema migrans (EM). This lesion
can progress to a bulls-eyetype lesion with the inner part clear and the edges
red. About 60% of cases have this sign. Other symptoms of the disease can be
fever, malaise, fatigue, myalgia, and migratory arthralgias. A wide range of
neurological symptoms have been reported, including facial palsy, aseptic men-
ingitis, chorea, myelitis, and encephalitis. Cardiac symptoms may occur a few
weeks to months after EM, including atrioventricular block. Rarely acute myo-
pericarditis or cardiomegaly have been reported. A chronic infection may lead to
arthritis in the large joints, polyneuropathy, or leukoencephalitis.
When Borrelia burgdorferi was first described, it was thought to be the only
agent of Lyme disease (15). Research in the last decade has indicated that at least
eight species of Borrelia have been found in the Ixodes vectors, four of which
have been demonstrated to cause human disease. All four species are known to
cause EM. B. burgdorferi is present in North America and Europe but is absent
from Russia and Asia. This species is most often associated with polyarthritis.
In Eurasia, Borrelia garinii is associated with neurological symptoms; it has
Tickborne Infections 405
been isolated from Japan to western Europe. Borrelia afzelii, another Eurasian
Borrelia, found from Japan and China to western Europe, is associated with the
chronic skin condition acrodermatitis chronica atrophicans. VS116 occurs in
Europe and has only been associated with EM (15a).
Though antibodies to B. burgdorferi are present in a significant fraction of
blood donors in endemic areas, no case of transfusion-transmitted Lyme disease
has been reported. B. burgdorferi has been cultured from the blood of patients
with early Lyme disease (16), which has led to the concern that Lyme disease may
be transmissible by transfusion (17). We have shown (18) that B. burgdorferi can
survive in stored blood products under frozen, refrigerated, or 2024C storage
conditions for the duration of the storage period of fresh frozen plasma (FFP), red
cells, and platelets, respectively. Because the characteristic EM rash is thought to
be caused by skin growth of spirochetes at the site of the tick bite, it is likely that
intravenously injected B. burgdorferi would not cause EM. Since the remainder
of the symptoms of Lyme disease are nonspecific, it is likely that, if transfusion-
transmitted Lyme disease exists, it would not be easily recognized.
Despite active surveillance for transfusion-transmitted Lyme disease, no
cases have been identified. However, we reported a case in which B. burgdorferi
DNA was detected in red cells and plasma from a donor who discovered EM the
day after blood donation and notified the blood center (19). He was confirmed
serologically to have Lyme disease. The involved blood products were cultured
but were negative for B. burgdorferi. Had this donor not called the blood center,
and had his blood products been transfused, it is not clear whether the recipients
would have been infected. It is also not clear whether the PCR-positive blood
units contained viable organisms.
In a case that illustrates the difficulty of transfusion transmission of Lyme
disease, we identified an implicated donor in a case of posttransfusion babesiosis.
Upon interview, the donor gave a history of having developed EM several days
following his blood donation and had been diagnosed and treated for Lyme
disease with a characteristic serial serological pattern for B. burgdorferi. The
recipient, who had a renal transplant and was on an immunosuppressive agent,
developed a typical serological response to babesiosis but did not seroconvert for
B. burgdorferi or become ill with Lyme disease (20). This was the case despite
the likelihood that both agents were transmitted to the donor by the bite of a
single, doubly infected, tick (21) 13 weeks before the blood donation. This case
suggests that the spirochetemic phase of B. burgdorferi after an infected tick bite
must be very short.
In an effort to estimate the risk of Lyme disease from transfusion, we
conducted serological follow-up on 155 open heart surgery patients who re-
ceived a total of 601 red cells and 371 platelet concentrates. None of the re-
cipients seroconverted for B. burgdorferi (95% CI, 00.5% for red cells and
00.8% for platelet concentrates) (22). At the same time, the study detected
406 Cable and Trouern-Trend
one case of seroconversion for babesiosis and placed the per unit risk of exposure
to babesiosis from a red cell transfusion in Connecticut at 0.17% (95% CI,
0.0040.9%).
VII. RICKETTSIAE
The rickettsia are aerobic gram-negative bacilli that are obligate intracellular par-
asites. Several genera (Rickettsia, Ehrlichia, Coxiella, and Orientia) are associ-
ated with human disease. Humans are most often accidental hosts. Infection
usually circulates in nature between animal hosts and arthropod vectors (ticks,
mites, fleas, lice). In some infections an asymptomatic rickettsemic stage may
precede symptoms. It is most likely during this period that rickettsial disease may
be transmitted by donated blood.
and some are serious veterinary pathogens. At least five species of Babesia or
Babesia-like organisms have been shown to infect humans, four with demonstra-
ble disease.
1. Babesia microti
Primarily a parasite of small rodents, particularly the white-footed deer mouse
(33), this species has caused several hundred infections in the United States,
primarily in the Northeast and upper Midwest (34). A significant number of these
cases have been asymptomatic. The vector Ixodes scapularis (Ixodes damini)
ranges from New Hampshire to Maryland, west to Wisconsin and Minnesota (14).
The ecology of B. microti infections in humans closely mirrors that of Lyme
disease and human granulocytic ehrlichiosis (HGE). Nymphs of the vector
I. scapularis bite humans and transmit the parasite. The ticks are infected as
larvae and acquire the infection from parasitic mice or voles. Deer, important to
the life cycle of I. scapularis and transmission of Lyme disease, are not infected
with B. microti and are not reservoirs for the parasite.
Infection follows a bite of an infected nymphal tick. This stage feeds
from May through September. The Babesia organism penetrates an erythrocyte.
The trophozoite multiplies by binary fission and forms tetrads. Lysis of the
erythrocyte releases the merozoites into the blood where they can reinfect other
cells, maintaining the infection or infecting a feeding tick. Transovarial passage
occurs in ticks allowing maintenance of the infection without a host (35).
Posttransfusion babesiosis is discussed below.
to 16% have been reported from northern California. Infections have occurred in
both splenectomized persons and those with intact spleens (37,38).
Blood
Case Age, gender, year Evidencea component Patient factors Donor infection Ref.
Until recently, all reported cases of transfusion babesiosis have been caused
by B. microti, which is endemic in New England, on Long Island, and in the upper
Midwest. Other recently reported species of Babesia and Babesia-like piroplasms
are discussed below.
The first case of transfusion babesiosis was reported in 1980 (44) (Table 2,
Case 1). The patient was a 70-year-old man who was on prednisone and
transfused in 1979 with 20 units of platelet concentrates for neutropenia, anemia,
and thrombocytopenia, diagnosed as ITP, then splenectomized 4 weeks later. Four
weeks after the splenectomy (8 weeks after the transfusion), he developed a fever
and was noted to have Babesia on a peripheral smear. He was resistant to
treatment with chloroquine and pentamidine and was treated successfully with
two exchange transfusions 6 weeks apart. This case is typical in that the patient
had had a splenectomy and was elderly and immunosuppressed. It was atypical
in that the only blood components received were platelet concentrates. However,
platelet concentrates can contain up to 0.5 mL of red cells.
By todays standards, the case for transfusion transmission in this first
report was not particularly compelling, since the only evidence for the alleged in-
fected donor, a summer resident of Nantucket, was an elevated Babesia serolog-
ical titer 3 months after donation. Hamster bioassay of a new sample from the
donor 3 months after the donation was negative. In addition, the incubation period
was 8 weeks, on the long side. A follow-up letter (49) correctly suggested that the
patient may have been already infected with unrecognized babesiosis and that the
splenectomy and/or prednisone, not the transfusion, may have been responsible
for the clinical observations. Although the patient had no history of a visit to
hyperendemic areas such as Nantucket or the New England off-shore islands,
much of coastal New England may have had Babesia-infected ticks. (Most
babesiosis patients do not report a specific tick bite.) However, the patient
seroconverted for Babesia antibody during the course of his illness, making
transfusion transmission more likely.
Confirmation of transfusion babesiosis requires the following evidence (50):
1. The patient must display clear-cut evidence of babesiosis with no other
source identified. Ideally the serologic and clinical evidence excludes
long standing subclinical infection.
2. In addition, a donor must be identified in the typical incubation period
of transfusion babesiosis who, at a minimum, has a history of travel or
residence in an endemic area and a positive Babesia antibody.
3. The donor should demonstrate recent infectivity by hamster inocula-
tion, PCR, or (less definitively) typical acute serologic progression.
For the purpose of analysis of case reports, we have used Possible to de-
scribe cases in which only criteria 1 are met, Probable for cases in which
criteria 1 and 2 are met, and Definite for cases in which all three criteria
are met.
414 Cable and Trouern-Trend
The first definite case of transfusion babesiosis was reported in 1982 (51)
from a transfusion in 1980 (Table 2, Case 3). The recipient was a premature
infant. The donor was a healthy summer resident of Fire Island (Long Island,
NY), who was demonstrated to be parasitemic by hamster inoculation a year after
the original exposure to infected ticks.
In 1994, stimulated by an increase of tickborne babesiosis in Connecticut
(52) and by three transfusion case reports to the Connecticut Red Cross Blood
Center, the authors initiated a registry of posttransfusion babesiosis cases. Blood
centers and Babesia researchers in New England, New York City, Long Island,
and Minnesota were contacted and asked to provide case reports. The reports
were tabulated in a standardized format and returned to the reporter for verifica-
tion. An update to this survey was conducted in 1996.
The data that follow are a summary of the data retained in this registry as
of July 1999. In total, 17 cases of transfusion-transmitted babesiosis have been
reported: 10 published and 7 unpublished. Table 2 is a summary of those cases,
along with references to the published cases.
Table 3 is a summary of patient factors revealed by these case reports.
Transfusion babesiosis occurs in all age groups and both sexes. There appears to
be a preponderance of older recipients, but this observation may merely reflect
the transfused population. Splenectomy, which is a risk factor for tickborne
disease, also appears to be a risk factor for transfusion transmission. Other
possible risk factorsprematurity, cancer and immunosuppressive chemotherapy,
and renal failuremay or may not be real, since these are also prevalent in
transfused patients. In any event, a significant proportion of cases appear to have
no complicating medical illness, with transfusion occurring in the context of
surgery or trauma.
The transfusion incubation period appears to be 28 weeks, with case 14
being an outlier. Case 14 illustrates the difficulty of identifying an implicated
donor. The case involved 49 donors, many of whom could not be contacted.
However, 22 were tested for Babesia antibody. Since the seroprevalence of
babesiosis in Connecticut is reported to be 1.02.6% (53), there is a high
likelihood of falsely implicating a donor not responsible for transmission. The ap-
parently implicated donor was seropositive and had a clear risk history. However,
he donated platelets by apheresis, which are relatively free of red cells, and the
apparent incubation period was 4.5 months, much longer than previously reported
in the other cases.
Transfusion babesiosis is a treatable disease, with the current recommended
therapy being clindamycin and quinine (43). Case 17 illustrates drug resistance
to clindamycin and quinine in a newborn, who subsequently responded to
atovoquone. Exchange transfusion to remove infected erythrocytes has also been
used successfully in the past (44). Outcome in general is favorable, with only one
death attributed to the infection among 19 patients.
Tickborne Infections 415
Early detection and a high index of suspicion are important in the manage-
ment of transfusion babesiosis. The disease is often missed, particularly in areas
not endemic for the disease. It is also confused with transfusion malaria, although
a skilled hematology technologist should be able to differentiate the intra-
erythrocytic parasites. Any patient who develops fever and a hemolytic anemia
after transfusion (otherwise not explained) should be suspected to have transfu-
sion babesiosis or malaria. They should be evaluated with review of a thick blood
smear by a hematology technologist experienced in such diagnoses.
Table 4 illustrates the geographic origin of implicated donors. Most were
from New England or Long Island, NY. The two cases from the Midwest reflect
the known endemic focus of infected ticks in this area. The predominance of cases
from Connecticut may, in fact, reflect the relatively high degree of exposure of
Connecticut blood donors. But it may also reflect an effort on the part of the blood
center and Connecticut health officials to educate physicians about babesiosis
(54). It is likely that a large number of cases are subclinical throughout the
endemic areas for babesiosis.
Implicated donors, in general, are asymptomatic and, typical of subclinical
babesiosis, often appear to have been infected for months to years. Some donors
have evidence for systemic disease, which may also reflect Lyme disease and
other tickborne illnesses (55).
Table 5 considers the blood components involved in the 17 reported cases.
Not surprisingly, most cases are transmitted by red cells. Platelets have been
implicated in only four of the reported cases, and in only one of these cases is
there definite implication of the involved donor.
In only one case (Case 17) has it been possible to evaluate co-components.
This interesting case illustrates the variable reactivity of red cells, since four other
patients besides the index case were transfused with other aliquots of the red cells
and only two were infected. The two uninfected red cell recipients were new-
borns, who received smaller doses of infected red cells (56) compared with the
three infected patients. A platelet concentrate made from the same donation did
not transmit Babesia to the recipient.
These cases also provide information on the survival of the Babesia parasite
under blood bank storage conditions. Definite cases of transfusion babesiosis
have been reported with red cells as old as 35 days and platelets as old as 4 days.
This is considerably longer than predicted from laboratory storage conditions
(48). However, in the laboratory, storage was in EDTA whole blood in glass tubes.
It is likely that blood bank storage conditions favoring red cell and/or platelet
survival would also be more favorable for the survival and infectivity of the
Babesia parasite.
The transfusion-transmission of Babesia species other than B. microti that
affect humans is to be expected. However, there has been no report of transfusion
transmission of B. divergens, the clinical babesiosis in Europe. Recently, two or
Tickborne Infections 417
three different human species in the United States have been describedin
Washington State (WA-1) (37,57), in Missouri (MO1) (39), and in northern
California (38). The reports of WA-1 infection have now been supplemented with
a report of a transfusion-transmitted case caused by a WA-1like organism (58),
the first transfusion babesiosis not caused by B. microti.
This case occurred in a multiply transfused, 76-year-old, spleen-intact man
with myelodysplasia and postcoronary artery bypass graft and aortic valve
replacement. Originally thought to have malaria, he was treated with chloroquine
and primaquine with initial resolution of symptoms. Later review at the Centers
for Disease Control and Prevention (CDC) identified the unusual piroplasm in his
red cells. The species was identified as very close to WA-1 serologically and
noncross-reactive with B. microti. His symptoms of fever and persistent parasite-
mia recurred, and he was treated with clindamycin and quinine; he appeared to
clear his parasitemia. Fifty-seven blood donors were investigated2 units of
whole blood, 15 red cells, 36 platelets, and 4 FFP. The implicated donor was a
34-year-old red cell donor with a titer against the recipients organism of 1:4096.
He lived in rural Washington State and was frequently exposed to deer. He was
parasitemic by hamster inoculation 7 months after his implicated donation.
No co-component or lookback donations could be evaluated because the recipi-
ents had died or were unavailable.
This case report is strikingly similar to many B. microti case reports:
misdiagnosis early (in this case, as malaria), chronic infection in both donor and
recipient, and a mild clinical course in the recipient. It cannot be determined from
the available case reports of tickborne illnesses and this transfusion case whether
any of the newly reported piroplasms will represent a significant public health
threat, either from tick transmission or blood transfusion. It is clear that apparent
transfusion babesiosis should be suspected outside of the endemic areas for
B. microti. The characteristic intraerythrocytic forms are the most reliable diag-
nostic tool. Serology to B. microti is likely to be negative or only weakly positive
in infections with these new organisms.
REFERENCES
1. AABB. FDA issues statement on blood donors possibly exposed to tick-borne
pathogens. AABB FAXNET 1997; No. 341.
1a. Arguin PM, Singleton J, Rotz LD, Marston E, Treadwell TA, Slater K, Chamberland
M, Schwartz A, Tengelsen L, Olson JG, Childs JE. An investigation into the
possibility of transmission of tick-borne pathogens via blood transfusion. Transfusion
1999; 39:828833.
2. Harwood RF, James MT. Entomology in Human and Animal Health. New York:
Macmillan Publishing Company, 1979:371378.
3. Krantz GW. A Manual of Acarology. 2nd ed. Corvalis: Oregon State University,
1878:211215.
4. Schmidt GD, Roberts LS. In: Foundations of Parasitology. 4th ed. St. Louis: Times
Mirror/Mosby College Publishing, 1989:660663.
5. Schmidt GD, Roberts LS. In: Foundations of Parasitology. 4th ed. St. Louis: Times
Mirror/Mosby College Publishing, 1989:671679.
6. Shulman IA, Appleman MD. An overview of unusual diseases transmitted by blood
transfusion within the United States. In: Westphal RG, Carlson KB, Turc JM, eds.
Emerging Global Patterns in Transfusion-Transmitted Infections. Arlington, VA:
American Association of Blood Banks, 1990:111.
7. Murray PR, Drew WL, Kobayashi GS, Thompson JH. Medical Microbiology. C.V.
St. Louis: Mosby, 1990:628632.
8. Monath TP, Tsai TF. Flaviviruses. In: Richman DD, Whitley RJ, Hayden FG, eds.
Clinical Virology. New York: Churchill Livingston, 1997:11331134.
9. Wahlberg P, Saikku P, Brummer-Korvenkontio M. Tick-borne viral encephalitis in
Finland. The clinical features of Kumlinge disease during 19591987. J Intern Med
1989; 225(3):173177.
10. Costero A, Grayson MA. Experimental transmission of Powassan virus (Flaviviridae)
by Ixodes scapularis ticks (Acari:Ixodadae). Am J Trop Med Hyg 1996; 55(5):536546.
11. Telford SR 3rd, Armstrong PM, Katavolos P, Foppa I, Garcia AS, Wilson ML,
Spielman A. A new tick-borne encephalitis-like virus infecting New England deer
ticks, Ixodes dammini. Emerg Infect Dis 1997; 3(2):165170.
12. Wong JK. Colorado tick fever virus and other arthropod-borne Reoviridae. In:
Richman DD, Whitley RJ, Hayden FG, eds. Clinical Virology. New York: Churchill
Livingston, 1997:755764.
13. Transmission of Colorado tick fever virus by blood transfusion. MMWR 1975;
24:425427.
14. CDC. Lyme diseaseUnited States, 1996. MMWR 1997; 46(23):531535.
15. Johnson RC, Schmid GP, Hyde FW, Steigerwalt AG, Brenner DJ. Borrelia burg-
dorferi sp. nov.: etiological agent of Lyme disease. Int J System Bacteriol 1984;
34:496497.
15a. Baranton G, Postic D, Saint Girons I, Boerlin P, Piffaretti JC, Assous M, Grimont PA.
Delineation of Borrelia burgdorferi sensu stricto, Borrelia garinii sp. nov. and group
VS461 associated with Lyme borreliosis. Int J System Bacteriol 1992; 42:378383.
420 Cable and Trouern-Trend
16. Benach JL, Bosler EM, Hanrahan JP. Spirochetes isolated from the blood of two
patients with Lyme disease. N Engl J Med 1983; 308:740742.
17. Aoki SK, Holland PV. Lyme diseaseanother transfusion risk? Transfusion 1989;
29:646650.
18. Badon SJ, Fister RD, Cable RG. Survival of Borrelia burgdorferi in blood products.
Transfusion 1989; 29:581583.
19. Badon SJ, Cable RG, Morse EE, Aslanzadeh J, Malawista SE. Evidence of Borrelia
burgdorferi in a blood donation. Blood 1995; 86(suppl 1):355a.
20. Cable RG, Krause P, Badon S, Silver H, Ryan R. Acute blood donor co-infection with
Babesia microti (Bm) and Borrelia burgdorferi (Bb) (abstr). Transfusion 1993;
33(suppl):50s.
21. Piesman J, Hicks TC, Sinsky RJ, Obiri G. Simultaneous transmission of Borrelia
burgdorferi and Babesia microti by individual nymphal Ixodes dammini ticks. J Clin
Microbiol 1987; 25:20122013.
22. Gerber MA, Shapiro ED, Krause PJ, Cable RG, Badon SJ, Ryan RW. The risk of
acquiring Lyme disease or babesiosis from a blood transfusion. J Infect Dis 1994;
170:231234.
23. Harwood RF, James MT. Entomology in Human and Animal Health. New York:
Macmillan Publishing Company, 1979:400403.
24. Boyer KM, Munford RS, Maupin GO, et al. Tick-borne relapsing fever: an interstate
outbreak originating at Grand Canyon National Park. Am J Epidemiol 1977; 105(5):
469479.
25. Pratt HD, Darsie RF. Highlights in medical entomology in 1974. Bull Entomol Soc
Am 1975; 21:173176.
26. Hira PR, Husein SF. Some transfusion-induced parasitic infections in Zambia. J Hyg
Epidemiol Microbiol Immunol 1979; 23(4):436444.
27. Wang CW, Lee CU. Malaria and relapsing fever following blood transfusion. Chin
Med J 1936; 50:241.
28. Wells GM, Woodward TE, Fiset P, Hornick RB. Rocky Mountain spotted fever
caused by blood transfusion. JAMA 1978; 239(26):27632765.
29. Benenson AS, ed. Control of Communicable Diseases Manual. 16th ed. Washington,
DC: American Public Health Association, 1995:392395.
30. Casleton BG, Salata K, Dasch GA, Strickman D, Kelly DJ. Recovery and viability
of Orientia tsutsugamushi from packed red cells and the danger of acquiring scrub
typhus from blood transfusion. Transfusion 1998; 38:680689.
31. Walker DH, Dumler JS. Emergence of the ehrlichioses as human health problems.
Emerg Infect Dis 1996; 2(1):1829.
32. Magnarelli LA, Dumler JS, Anderson JF, Johnson RC, Fikrig E. Coexistence of
antibodies to tick-borne pathogens of babesiosis, ehrlichiosis and Lyme borreliosis in
human sera. J Clin Microbiol 1995; 33:30543057.
32a. Eastlund T, Persing D, Mathiesen D. Human granulocytic ehrlichiosis after red cell
transfusion. Transfusion 1999; 39(suppl):117s.
33. Spielman A, Etkind P, Piesman J. Reservoir hosts of human babesiosis on Nantucket
Island. Am J Trop Med Hyg 1981; 30:560565.
34. Dammin GJ, Spielman A, Benach JL. The rising incidence of clinical Babesia microti
infection. Hum Pathol 1981; 12:398400.
Tickborne Infections 421
35. Murray PR, Drew WL, Kobayashi GS, Thompson JH. Medical Microbiology.
St. Louis: Mosbey, 1990:375377.
36. Gelfand JA. Babesia. In: Mandell GL, Bennett JE, Dolin RA, eds. Principles and
Practice of Infectious Diseases. 4th ed. New York: Churchill Livingston, 1995:2497
2500.
37. Quick RE, Herwaldt BL, Thomford JW, Garnett ME, Eberhard ML, Wilson M, Spach
DH, Dickerson JW, Telford SR 3d, Steingart KR. Babesiosis in Washington State: a
new species of Babesia? Ann Intern Med 1993; 119(4):284290.
38. Persing DH, Herwaldt BL, Glaser C, Lane RS, Thomford JW, Mathiesen D, Krause
PJ, Phillip DF, Conrad PA. Infection with a Babesia-like organism in northern
California. N Engl J Med 1995; 332(5):298303.
39. Herwaldt B, Persing DH, Prcigout EA, Goff WL, Mathiesen DA, Taylor PW,
Eberhard ML, Gorenflot AF. A fatal case of babesiosis in Missouri: identification of
another piroplasm that infects humans. Ann Intern Med 1996; 124(7):643650.
40. Shih CM, Liu LP, Chung WC, Ong SJ, Wang CC. Human babesiosis in Taiwan:
asymptomatic infection with a Babesia microti-like organism in a Taiwanese woman.
J Clin Microbiol 1997; 35(2):450454.
41. Ruebush TK II, Juranek DD, Spielman A. Epidemiology of human babesiosis on
Nantucket Island. Am J Trop Med Hyg 1981; 30:937941.
42. Benach JL, Habicht GS. Clinical characteristics of human babesiosis. J Infect Dis
1981; 144:481.
43. Clindamycin and quinine treatment for Babesia microti infections. MMWR 1983;
32:6572.
44. Jacoby GA, Hunt JV, Kosinski KS, Demirjian ZN, Huggins C, Etkind P, Marcus LC,
Spielman A. Treatment of transfusion-transmitted babesiosis by exchange transfu-
sion. N Engl J Med 1980; 303:10981100.
45. Popovsky MA, Lindberg LE, Syrek AL, Page, PL. Prevalence of Babesia antibody
in a selected blood donor population. Transfusion 1988; 28:2961.
46. Benach JL, Coleman JL, Habicht GS. Serological evidence for simultaneous occuren-
ces of Lyme disease and babesiosis. J Infect Dis 1985; 152:473477.
47. Krause PJ, Spielman A, Telford SR III, Sikand VK, McKay K, Christianson D,
Pollack RJ, Brassard P, Magera J, Ryan R, Persing EH. Persistent parasitemia after
acute babesiosis. N Engl J Med 1998; 339:160165.
48. Eberhard ML, Walker EM, Steurer F. Survival and infectivity of Babesia in blood
maintained at 25C and 2-4C. J Parasitol 1995; 81:790792.
49. Przyjemski CJ. Was babesiosis transmitted by transfusion? N Engl J Med 1981;
304:733.
50. Badon SJ, Cable RG. Investigation of suspected cases of post-transfusion babesiosis
(abstr). Transfusion 1997; 37(suppl):56S.
51. Wittner M, Rowin KS, Tanowitz HB, Hobbs JF, Saltzman S, Wenz B, Hirsch R,
Chisholm E, Healy GR. Successful chemotherapy of transfusion babesiosis. Ann
Intern Med 1982; 96:601604.
52. State of Connecticut Department of Health Services. BabesiosisConnecticut 1990
1996. Conn Epidemiol 1997; 17:1819.
53. Krause PJ, Telford SR, Ryan R. Geographic and temporal distribution of Babesial
infection in Connecticut. J Clin Microbiol 1991; 29:14.
422 Cable and Trouern-Trend
18
Surveillance for
Transfusion-Transmitted
Infectious Diseases
Mary E. Chamberland and Rima F. Khabbaz
Centers for Disease Control and Prevention, Atlanta, Georgia
I. INTRODUCTION
The U.S. blood supply is among the safest in the world. The risk of transfusion-
transmitted infections has been markedly reduced as a result of improvements in
donor screening and serological testing, the development and implementation of
viral inactivation procedures for plasma-derived products, and changes in trans-
fusion practices. Nevertheless, since blood is a human tissue, it is a natural vehicle
for transmission of infectious agents. The transmission of human immunodefi-
ciency virus (HIV) through the blood supply in the early 1980s and the more
recent experiences with hepatitis C virus (HCV) transmission from intravenous
immune globulin (IVIG) (1) and bacterial sepsis from contaminated albumin (2)
point to the need for continued vigilance regarding both known and unrecognized
or uncharacterized threats to the blood supply.
An important component of the multifaceted approach to help ensure
the safety of the blood supply is surveillance. As part of its retrospective re-
view of the events of the early 1980s that led to the transmission of HIV
through blood and blood products, the Institute of Medicine developed a se-
ries of recommendations to help thwart future threats to the blood supply. One
of these was a recommendation to establish a surveillance system . . . [to] de-
tect, monitor, and warn of adverse effects in recipients of blood and blood
products (3).
423
424 Chamberland and Khabbaz
Figure 1 Adult and pediatric cases of acquired immunodeficiency syndrome in the United States attributed to receipt of blood transfusion,
Chamberland and Khabbaz
Figure 2 Adult and pediatric cases of acquired immunodeficiency syndrome in the United States among persons with hemophilia or other
427
Figure 3 HIV seroprevalence in blood donors in the United States, by date of donation, November 1985December 1993. (From Ref. 10.)
Chamberland and Khabbaz
Surveillance for Transfusion-Transmitted Infectious Diseases 429
infections reported worldwide is small, and most have occurred among persons
in West and Central Africa countries (20). However, because these infections are
inconsistently detected by current enzyme immunoassays (EIAs) for antibodies
to HIV-1, there are important implications for blood safety (21). CDC has an
ongoing program to evaluate HIV-seropositive blood donors for unusual variants;
previous studies that evaluated blood donors and other populations have not found
any evidence of infection with HIV-1 group O (22). In addition, CDC has
established active international and domestic surveillance for divergent HIV
strains to monitor their prevalence, evaluate the sensitivity of licensed HIV
serological tests for detecting these variants, and assist in the modification of
screening tests to improve their sensitivity. As part of this surveillance program,
CDC, in collaboration with state health departments, is undertaking epidemiolog-
ical and laboratory investigations of persons who were reported with HIV/AIDS
and who were born in a country where HIV-1 group O has been documented (i.e.,
West and Central Africa). Two HIV-1 group O infections among persons in the
United States have been reported through this sentinel surveillance; both persons
were originally from Africa, and neither was a blood donor (23,24). Efforts are
underway to modify existing HIV-EIAs to improve detection of group O strains,
without compromising sensitivity for the group M viruses. As an interim measure,
the U.S. Food and Drug Administration (FDA) has recommended that donors at
increased risk for HIV-1 group O infection (e.g., resident in areas where group O
strains are endemic) be deferred from donating blood or plasma.
D. HIV-2
HIV-2, another retrovirus that causes AIDS and is endemic in many countries in
West Africa, has been transmitted by transfusion of blood and blood products
in Europe (25). Epidemiological and donor testing data indicate that the preva-
lence of HIV-2 among blood donors in the United States is very low. Neverthe-
less, because of the potential for transmission by blood and blood products and
the availability of licensed combination HIV-1/HIV-2 EIAs, FDA recommended
in June 1992 that blood and plasma donors be screened for antibodies to HIV-2.
No cases of transfusion-acquired HIV-2 infection have been reported in the
United States, and only three HIV-2 seropositive units have been found among
whole blood and plasma donors (26).
Figure 4 Cases of hepatitis B and hepatitis C/non-A, non-B hepatitis attributed to blood transfusion from selected states, by year of report,
19831995. (From Ref. 27.)
Chamberland and Khabbaz
Surveillance for Transfusion-Transmitted Infectious Diseases 433
days; range 54192 days) during which antibodies cannot be detected; and an
inability to reliably detect chronic infection (15,32). A third-version anti-HCV
test, recently approved for screening blood donors, may further improve the
ability to detect anti-HCV antibodies in a low-prevalence population, such as
blood donors (33).
Although viral inactivation procedures have virtually eliminated the risk of
HCV infection from plasma-derived products, the first outbreak of hepatitis C
associated with contaminated IVIG was reported in the United States in 1994 (1).
Infection was associated with lots of Gammagard (Baxter Healthcare Corpora-
tion, Deerfield, IL) produced from plasma screened by second-version anti-HCV
assays that were positive for HCV RNA. It has been hypothesized that the
second-version tests removed most of the complexing antibodies from plasma
used to make IGIV, ultimately resulting in more HCV in the IG fraction than in
the non-IG fraction. In addition, manufacture of this particular product did not
include a viral inactivation step as a further measure for product safety.
donors and screening test sensitivity) (36) to one in 641,000 (based on a study of
incident infections among repeat blood donors in the infectious window period)
(15). The former estimate is similar to that observed in a prospective study by
Nelson and coworkers of patients who received transfusions during cardiac
surgery between 1985 and 1991 (37). They found that the combined rate of
transfusion-transmitted HTLV-I and -II fell from about one in 8,500 units trans-
fused before donor screening to one in 69,272 units after institution of screening.
The relative insensitivity of combined tests, particularly in detecting HTLV-II (the
more common retrovirus of the two in the United States), has been a concern.
Although as many as 43% of HTLV-IIinfected donations may have gone
undetected when earlier EIAs were used (38), more recent assays have reduced
this proportion to about 34% (39).
V. CREUTZFELDT-JAKOB DISEASE
Creutzfeldt-Jakob disease (CJD) is a rare, invariably fatal neurodegenerative
disease believed to be caused by an unconventional, disinfection-resistant infec-
tious agent (40). The likely causative agent is a prion protein, which is a
confirmationally altered form of a normal plasma membrane protein. Iatrogenic
cases of CJD in recipients of human pituitary-derived growth hormone and dura
mater grafts have had incubation periods of as long as 20 years. The CJD agent
has been reported occasionally in the blood of CJD patients at low titer, and
inoculation of peripheral blood mononuclear cells and some derivatives of plasma
from CJD patients into rodent brains has resulted in CJD-like lesions (4043).
In contrast to these experimental data, there has been to date no confirmed reports
of CJD resulting from receipt of blood transfusion. Epidemiological case-control
studies show no increase in the frequency of transfusions among CJD patients
compared with controls (4447).
CDC has several systems of surveillance in place to help assess the risk, if
any, of transmission of CJD by transfusion of blood and blood products. CDC
conducts routine surveillance for CJD through ongoing review of national mor-
tality data. Results from 1979 through 1994 indicate that annual rates of CJD have
remained stable (at about one case per million population) (48). Thus, despite
regular blood donation by persons who subsequently develop CJD, blood trans-
fusions do not appear to be amplifying CJD infections in the U.S. population.
None of the 3642 reported cases of CJD was also reported to have hemophilia,
thalassemia, or sickle cell diseasediseases associated with increased exposure
to blood or blood products, such as clotting factor concentrates and/or cryopre-
cipitate or red blood cells (49). Two studies suggest that routine mortality
surveillance has good sensitivity to detect CJD cases. The first study, conducted
in 11 states, found that 80% of all neuropathologically confirmed cases of CJD
Surveillance for Transfusion-Transmitted Infectious Diseases 435
B. Chagas Disease
Chagas disease, a vectorborne disease caused by the parasite Trypanosoma cruzi,
is endemic in parts of Central and South America and Mexico. Transmission in
nature occurs by cutaneous or mucosal contact with the feces of reduvid bugs.
If untreated, Chagas disease can result in lifelong, asymptomatic parasitemia;
infected individuals can in turn transmit T. cruzi through transfusion. Receipt of
blood transfusions is the most common means of transmission in some disease-
endemic areas (66).
The immigration of thousands of persons from T. cruziendemic areas and
increased international travel have raised concerns about the increased potential
for transfusion-transmitted Chagas disease in North America (54). It has been
estimated that at least 100,000 persons with chronic T. cruzi infection reside in
the United States (66). To date, there have been three reported cases of Chagas
disease from transfusions in the United States and one in Canada; all four patients
were immunocompromised and developed acute, symptomatic disease, which
facilitated their recognition (54,66). The American National Red Cross has
conducted limited lookback studies of recipients of blood and blood products that
were seropositive for antibodies to T. cruzi and found no evidence of transmis-
sion, suggesting that the transmission rate in the United States may be no higher
than 10% (58,67). Until recently, reliable seroprevalence estimates among blood
donors have been hampered by the lack of serological tests with adequate
sensitivity and specificity. More recent studies using screening and confirmatory
testing schemas and conducted among selected blood donors who were more
likely to be at increased risk for T. cruzi infection because of birth in or travel to
disease-endemic countries have found approximately 0.1% of such donors to be
seropositive for antibodies to T. cruzi (58,6769). Extrapolating from these and
other studies, Dodd estimated that the overall risk for T. cruzi contamination is
one in 42,000 per unit of donated blood in the United States (58). Although there
is no national surveillance system in place to monitor transfusion-associated
Chagas disease, the drug (nifurtimox) that is used to treat acute infection in the
United States is available only through CDCs Drug Service. Hence, CDC would
likely learn of diagnosed, symptomatic cases of acute infection.
C. Leishmaniasis
Leishmaniasis is a parasitic disease, primarily found in the tropics and subtropics,
that is transmitted in nature by the bite of infected sand flies. Leishmaniasis can
cause skin lesions (cutaneous leishmaniasis) or mucosal lesions (mucosal leish-
maniasis) or invade internal organs (e.g., liver, spleen, bone marrow) of the body
(visceral leishmaniasis) and cause anemia, weakness, and sometimes death,
if untreated. Visceral leishmaniasis, caused by organisms in the Leishmania
438 Chamberland and Khabbaz
D. Malaria
Malaria is a reportable disease in the United States, and cases confirmed by blood
smear are reported to local and/or state health departments by health care
providers and/or laboratories (74). CDC also directly obtains reports of other
cases from health care providers who request assistance in the diagnosis or
treatment of malaria. All reported cases of malaria that are acquired in the United
States are fully investigated to collect detained clinical and epidemiological data.
For cases in persons who report no history of international travel but who received
a blood transfusion, the blood bank is contacted to identify and collect blood
specimens from potentially implicated donors. Donors are serologically tested,
Surveillance for Transfusion-Transmitted Infectious Diseases 439
and any seropositive donors are investigated. Implicated donors are more likely
to be identified by indirect fluorescent-antibody detection than by blood smear
examination alone; most have subpatent parasitemia, and a positive blood smear
is found only in approximately 30% of donors (75).
On average, approximately three cases per year of transfusion-induced
malaria are reported in the United States (75). The overall incidence of transfu-
sion-transmitted malaria in the United States is about one case per every 4 million
units of blood collected (75). The infecting Plasmodium species for the 99 cases
of transfusion-induced malaria reported in 19581994 were: P. malariae, 32
(32%); P. falciparum, 31 (31%); P. vivax, 28 (28%); P. ovale, 7 (7%); and
P. falciparum and P. malariae, 1 (1%) (M. Parise, personal communication).
Of the 70 implicated donors, 39 (56%) were foreign nationals (M. Parise,
personal communication). At present, the optimal means of preventing transmis-
sion of malaria by blood transfusion is through deferral of donors with a history
of malaria or a relevant travel history. CDCs investigations of transfusion-
acquired malaria cases indicate that most commonly implicated donors incor-
rectly answer questions regarding travel to a malaria-endemic area.
20,000 persons with hemophilia and related congenital blood clotting disorders
in the United States. Data about the nature and extent of joint and infectious
disease complications are collected in all 144 federally sponsored, specialized
hemophilia treatment centers throughout the country. In addition, a serum speci-
men is sent to CDC for serological testing for hepatitis and HIV infection and for
storage in a national serum bank for use in future investigations related to blood
safety issues. The system also has an acute illnessreporting system that facilitates
identification and investigation of potential infection sources and outbreaks and
the development of intervention strategies to prevent further disease occurrence.
IX. CONCLUSION
Since blood is a biological product, it is unlikely that the risk of transfusion-
transmitted infections will ever be reduced to zero. Nonetheless, the blood supply
in the United States in the 1990s is among the safest in the world and is safer now
than it has ever been. Factors contributing to the reduced risk of transfusion-
transmitted infections include improvements in donor selection, donor screening
by serological tests and other markers of infection, and viral inactivation proce-
dures for plasma-derived products. These approaches, coupled with surveillance
to monitor for trends in infection, are the mainstay of public health strategies to
protect the blood supply from known pathogens and to monitor for the emergence
of new infectious agents. CDC views surveillance as a critical component of its
overall mission. One of the four goals of CDCs strategic plan to help address
emerging infectious threats is the improvement and expansion of surveillance
capabilities for infectious diseases in the United States and internationally (76).
Enhanced surveillance can play an important role in helping to ensure the safety
of blood and plasma products.
REFERENCES
1. Bresee JS, Mast EE, Coleman PJ, Baron MJ, Schonberger LB, Alter MJ, Jonas MM,
Yu MW, Renzi PM, Schneider LC. Hepatitis C virus infection associated with admin-
istration of intravenous immune globulin. A cohort study. JAMA 1996; 276:1563
1567.
2. Centers for Disease Control and Prevention. Bacterial sepsis associated with receipt
of albumin. MMWR 1996; 45:866867.
3. Committee to Study HIV Transmission Through Blood and Blood Products, Division
of Health Promotion and Disease Prevention, Institute of Medicine. Leveton LB, Sox
HC Jr, Stoto MA, eds. HIV and the Blood Supply. An analysis of crisis decisionmak-
ing. Washington, DC: National Academy Press, 1995.
4. Thacker SB, Berkelman RL, Stroup DF. The science of public health surveillance.
J Public Health Policy 1989; 10:187203.
Surveillance for Transfusion-Transmitted Infectious Diseases 441
5. Thacker SB, Berkelman RL. Public health surveillance in the United States.
Epidemiol Rev 1988; 10:164190.
6. Centers for Disease Control and Prevention. HIV/AIDS Surveillance Report 1996;
8(no. 2):139.
7. Buehler JW, Berkelman RL, Stehr-Green JK. The completeness of AIDS surveillance.
J AIDS 1992; 5:257264.
8. Rosenblum L, Buehler JW, Morgan MW, Costa S, Hidalgo J, Holmes R, Lieb L,
Shields A, Whyte BM. The completeness of AIDS case reporting, 1988: A multistate
collaborative surveillance project. Am J Public Health 1992; 82:14951499.
9. Petersen LR, Dodd R, Dondero TJ Jr. Methodologic approaches to surveillance of
HIV infection among blood donors. Pub Health Rep 1990; 105:153157.
10. Centers for Disease Control and Prevention. National HIV Serosurveillance Sum-
mary: Update1993. Vol 3. Atlanta: U.S. Department of Health and Human Ser-
vices, 1995.
11. Centers for Disease Control and Prevention. National HIV Serosurveillance Sum-
mary: Results through 1992. Vol 3. Atlanta: U.S. Department of Health and Human
Services, 1993.
12. Johnson ES, Doll LS, Satten GA, Lenes B, Shafer AW, Kamel H, Casanova RJ,
Petersen LR. Direct oral questions to blood donors: the impact on screening for
human immunodeficiency virus. Transfusion 1994; 34:769774.
13. Lackritz EM, Satten GA, Aberle-Grasse J, Dodd RY, Raimondi VP, Janssen RS,
Lewis WF, Notari EP, Petersen LR. Estimated risk of transmission of the human
immunodeficiency virus by screened blood in the United States. N Engl J Med 1995;
333:17211725.
14. Zuck TF, Thomson RA, Schreiber GB, Gilcher RO, Kleinman SH, Murphy EL,
Ownby HE, Williams AE, Busch MP, Smith JW, Nass CC, Hollingsworth CG, Nemo
GJ for the REDS Group. The Retrovirus Epidemiology Donor Study (REDS);
rationale and methods. Transfusion 1995; 35:944951.
15. Schreiber GB, Busch MP, Kleinman SH, Korelitz JJ, for the Retrovirus Epidemiology
Donor Study. The risk of transfusion-transmitted viral infections. N Engl J Med 1996;
334:16851690.
16. Busch MP, Lee LLL, Satten GA, Henrard DR, Farzadegan H, Nelson KE, Read S,
Dodd RY, Petersen LR. Time course of detection of viral and serologic markers
preceding human immunodeficiency virus type 1 seroconversion: implications for
screening of blood and tissue donors. Transfusion 1995; 35:9197.
17. Lackritz EM, Stramer SL, Jacobs TA, Strauss D, Busch MP, Schable CA. Results of
national testing of U.S. blood donations for HIV-1 p24 antigen [Abstract 751]. In:
Program and Abstracts of the 4th Conference on Retroviruses and Opportunistic
Infections, Washington DC, January 2226, 1997.
18. Doll LS, Petersen LR, White CR, Ward JW, HIV Blood Donor Study Group. Human
immunodeficiency virus type 1-infected blood donors: behavioral characteristics and
reasons for donation. Transfusion 1991; 31:704709.
19. Williams AE, Thomson RA, Schreiber GB, Watanabe K, Bethel J, Lo A, Kleinman
SH, Hollingsworth CG, Nemo GJ, for the Retrovirus Epidemiology Donor Study.
Estimates of infectious disease risk factors in US blood donors. JAMA 1997;
277:967972.
442 Chamberland and Khabbaz
20. Hu DJ, Dondero TJ, Rayfield MA, George JR, Schochetman G, Jaffe HW, Luo C-C,
Kalish ML, Weniger BG, Pau C-P, Schable CA, Curran JW. The emerging genetic
diversity of HIV. The importance of global surveillance for diagnostics, research and
prevention. JAMA 1996; 275:210216.
21. Schable C, Zekeng L, Pau C-P, Hu D, Kaptue L, Gurtler L, Dondero T, Tsague J-M,
Schochetman G, Jaffe H, George JR. Sensitivity of United States HIV antibody tests
for detection of HIV-1 group O infections. Lancet 1994; 344:13331334.
22. Pau CP, Hu DJ, Spruill C, Schable C, Lackritz E, Kai M, George JR, Rayfield MA,
Dondero TJ, Williams AE, Busch MP, Brown AE, McCutchan FE, Schochetman G.
Surveillance for human immunodeficiency virus type 1 group O infections in the
United States. Transfusion 1996; 36:398400.
23. Sullivan PS, Jones J, Schable CA, Schochetman G, Pau C, Rayfield M, Do A, Tetteh
C, Ward JW. National surveillance for HIV-1 group O infections: Preliminary results
[Abstract 159]. In: Program and Abstracts of the 4th Conference on Retroviruses and
Opportunistic Infections, Washington, DC, January 2226, 1997.
24. Sullivan PS, Do AN, Robbins K, Kalish M, Subbarao S, Pieniazek D, Schable C,
Afaq G, Markowitz J, Myers R, Joseph JM, Benjamin G. Surveillance for variant
strains of HIV: subtype G and group O HIV-1 [letter]. JAMA 1997; 278:292.
25. OBrien TR, George JR, Holmberg SD. Human immunodeficiency virus type 2
infection in the United States: epidemiology, diagnosis, and public health importance.
JAMA 1992; 267:27752779.
26. Sullivan M, Williams A, Guido E, Metler R, Schable C, Stramer S. Detection and
characterization of an HIV type 2 antibody positive blood donor in the U.S. [abst
S229]. Transfusion 1997; 37(suppl):58S.
27. Centers for Disease Control and Prevention. Hepatitis Surveillance Report No. 56.
Atlanta: U.S. Department of Health and Human Services, 1995.
28. Alter MJ, Hadler SC, Margolis HS, Alexander WJ, Hu PY, Judson FN, Mares A,
Miller JK, Moyer LA. The changing epidemiology of hepatitis B in the United States.
Need for alternative vaccination strategies. JAMA 1990; 263:12181222.
29. Alter MJ, Mares A, Hadler SC, Maynard JE. The effect of underreporting on the
apparent incidence and epidemiology of acute viral hepatitis. Am J Epidemiol 1987;
125:133139.
30. Centers for Disease Control. Public Health Service inter-agency guidelines for
screening donors of blood, plasma, organs, tissues, and semen for evidence of
hepatitis B and hepatitis C. MMWR 1991; 40 (RR-4):117.
31. Alter MJ, Hadler SC, Judson FJ, Mares A, Alexander WJ, Hu PY, Miller JK, Moyer
LA, Fields HA, Bradley DW, Margolis HS. Risk factors for acute non-A, non-B
hepatitis in the United States and association with hepatitis C virus infection. JAMA
1990; 264:22312235.
32. Alter MJ. Residual Risk of Transfusion-Associated Hepatitis. NIH Consensus Devel-
opment Conference on Infectious Disease Testing for Blood Transfusions. Bethesda,
National Institutes of Health, 1995.
33. Gretch D. Diagnostic tests for hepatitis C. In Program and Abstracts of NIH
Consensus Development Conference on Management of Hepatitis C. Bethesda,
National Institutes of Health, 1997.
34. Khabbaz RF, Heneine W, Kaplan JE. Testing for other human retroviruses: HTLV-I
Surveillance for Transfusion-Transmitted Infectious Diseases 443
and HTLV-II. In: Schochetman G, George JR, eds. AIDS Testing. 2d ed. New York:
Springer-Verlag, 1994:206223.
35. Centers for Disease Control. Human T-lymphotropic virus type I screening in
volunteer blood donorsUnited States, 1989. MMWR 1990; 39:915, 921924.
36. Dodd RY. The risk of transfusion-transmitted infection [editorial]. N Engl J Med
1992; 327:419421.
37. Nelson KE, Donahue JG, Munoz A, Cohen ND, Ness PM, Teague A, Stambolis VA,
Yawn DH, Callicott B, McAllister H, Reitz BA, Lee H, Farzadegan H, Hollingsworth
CG. Transmission of retroviruses from seronegative donors by transfusion during
cardiac surgery: a multicenter study of HIV-1 and HTLV-I/II infections. Ann Intern
Med 1992; 117:554559.
38. Hjelle B, Wilson C, Cyrus S, Bradshaw P, Lo J, Schammel C, Wiltbank T, Alexander
S. Human T-cell leukemia virus type II infection frequently goes undetected in
contemporary U.S. blood donors. Blood 1993; 81:16411644.
39. Gallo D, Yeh ET, Moore ES, Hanson CV. Comparison of four enzyme immunoassays
for detection of human T-cell lymphotropic virus type 2 antibodies. J Clin Microbiol
1996; 34:213215.
40. DeArmond SJ, Prusiner SP. Etiology and pathogenesis of prion diseases. Am J Pathol
1995; 146:785811.
41. Manuelidis EE, Kim JH, Mericangas JR, Maneulidis L. Transmission to animals of
Creutzfeld-Jakob disease from human blood [letter]. Lancet 1985; II:896897.
42. Tamai Y, Kojima H, Kitajima R, Taguchi F, Ohtani Y, Kawaguchi T, Miura S, Sata
M, Ishihara Y. Demonstration of the transmissible agent in tissue from a pregnant
woman with Creutzfeldt-Jakob disease [letter]. N Engl J Med 1992; 327:649.
43. Tateishi J. Transmission of Creutzfeldt-Jakob disease from human blood and urine
into mice [letter]. Lancet 1985; II:1074.
44. Davanipour Z, Alter M, Sobel E, Asher DM, Gajdusek DC. A case-control study of
Creutzfeldt-Jakob disease. Dietary risk factors. Am J Epidemiol 1985; 122:443451.
45. Esmonde TFG, Will RG, Slattery JM, Knight R, Harries-Jones R, DeSilva R,
Matthews WB. Creutzfeldt-Jakob disease and blood transfusion. Lancet 1993;
341:205207.
46. Kondo K, Kuroiwa Y. A case control study of Creutzfeldt-Jakob disease: association
with physical injuries. Ann Neurol 1982; 11:377381.
47. Little BW, Mastrianni J, DeHaven AL, Brown P, Goldfarb L, Gajdusek DC. The
epidemiology of Creutzfeldt-Jakob disease in eastern Pennsylvania [Abstract 614P].
Neurology 1993; 43:A316.
48. Holman RC, Khan AS, Belay ED, Schonberger LB. Creutzfeldt-Jakob disease in the
United States, 19791994: using national mortality data to assess the possible
occurrence of variant cases. Emerg Infect Dis 1996; 2:333337.
49. Schonberger LB, Sullivan M, Holman RC, Austin H, Dodd R, Khan A. Creutzfeldt-
Jakob Disease (CJD) and blood safety [Abstract PW 40-4]. In: Abstracts of Xth
International Congress of Virology, Jerusalem, Israel, August 1116, 1996.
50. Davanipour Z, Smoak C, Bohr T, Sobel E, Liwnicz B, Chang S. Death certificates:
an efficient source for ascertainment of Creutzfeldt-Jakob disease cases. Neuro-
epidemiology 1995; 14:16.
444 Chamberland and Khabbaz
51. Centers for Disease Control and Prevention. Surveillance for Creutzfeldt-Jakob
diseaseUnited States. MMWR 1996; 45:665668.
52. Sullivan MT, Schonberger LB, Kessler D, Williams AE, Dodd RY. Creutzfeldt-Jakob
disease (CJD) investigational lookback study [Abstract S6]. Transfusion 1997;
37(suppl):2S.
53. Heye N, Hensen S, Muller N. Creutzfeldt-Jakob disease and blood transfusion [letter].
Lancet 1994; 343:298299.
54. Sloand EM, Pitt E, Klein HG. Safety of the blood supply. JAMA 1995; 274:1368
1373.
55. Wagner SJ, Friedman LI, Dodd RY. Transfusion-associated bacterial sepsis. Clin
Micro Rev 1994; 7:290302.
56. Centers for Disease Control and Prevention. Red blood cell transfusions contami-
nated with Yersinia enterocoliticaUnited States, 19911996, and initiation of a
national study to detect bacteria-associated transfusion reactions. MMWR 1997;
46:553555.
57. Hoppe PA. Interim measures for detection of bacterially contaminated red cell
components [editorial]. Transfusion 1992; 32:199201.
58. U.S. General Accounting Office. Blood Supply. Transfusion-Associated Risks.
GAO/PEMD-97-2. Washington, DC: Government Printing Office, 1997.
59. Dodd RY. Adverse consequences of blood transfusion: quantitative risk estimates. In:
Nance ST, ed. Blood Supply Risks, Perceptions and Prospects for the Future.
Bethesda, MD: American Association of Blood Banks, 1994:124.
60. AuBuchon JP. Blood transfusion options. Improving outcomes and reducing costs.
Arch Pathol Lab Med 1997; 121:4047.
61. Gerber MA, Shapiro ED, Krause PJ, Cable RG, Badon SJ, Ryan RW. The risk of
acquiring Lyme disease or babesiosis from a blood transfusion. J Infect Dis 1994;
170:231234.
62. Herwaldt BL, Kjemtrup AM, Conrad PA, Barnes RC, Wilson M, McCarthy MG,
Sayers MH, Eberhard ML. Transfusion-transmitted babesiosis in Washington State:
first reported case caused by a WA1-type parasite. J Infect Dis 1997; 175:12591262.
63. Mintz ED, Anderson JF, Cable RG, Hadler JL. Transfusion-transmitted bebesiosis: a
case report from a new endemic area. Transfusion 1991; 31:365368.
64. Eberhard ML, Walker EM, Steurer FJ. Survival and infectivity of Babesia in blood
maintained at 25C and 24C. J Parasitol 1995; 81:790792.
65. Popovsky MA. Transfusion-transmitted babesiosis [editorial]. Transfusion 1991;
31:296298.
66. Schmunis GA. Trypanosoma cruzi, the etiologic agent of Chagas disease: status in
the blood supply in endemic and nonendemic countries. Transfusion 1991; 31:547
557.
67. Leiby DA, Read EJ, Lenes BA, Yund AJ, Stumpf RJ, Kirchoff LV, Dodd RY.
Seroepidemiology of Trypanosoma cruzi, etiologic agent of Chagas disease, in US
blood donors. J Infect Dis 1997; 176:10471052.
68. Brashear RJ, Winkler MA, Schur JD, Lee H, Burczak JD, Hall HJ, Pan AA. Detection
of antibodies to Trypanosoma cruzi among blood donors in the southwestern and
western United States. I. Evaluation of the sensitivity and specificity of an enzyme
immunoassay for detecting antibodies to T cruzi. Transfusion 1995; 35:213218.
Surveillance for Transfusion-Transmitted Infectious Diseases 445
69. Schulman IA, Appleman MD, Saxena S, Hiti AL, Kirchhoff LV. Specific antibodies
to Trypanosoma cruzi among blood donors in Los Angeles, California. Transfusion
1997; 37:727731.
70. Magill AJ, Grogl M, Gasser RA Jr, Sun W, Oster CN. Visceral infection caused by
Leishmania tropica in veterans of Operation Desert Storm. N Engl J Med 1993;
328:13831387.
71. Magill AJ, Grogl M, Johnson SC, Gasser RA. Visceral infection due to Leishmania
tropica in a veteran of Operation Desert Storm who presented 2 years after leaving
Saudi Arabia. Clin Infect Dis 1994; 19:805806.
72. Grogl M, Daugirda JL, Hoover DL, Magill AJ, Berman JD. Survivability and
infectivity of viscerotropic Leishmania tropica from Operation Desert Storm partic-
ipants in human blood products maintained under blood bank conditions. Am J Trop
Med Hyg 1993; 49:308315.
73. Gunby P. Desert Storm veterans now may donate blood; others called for discussion
of donor tests. JAMA 1993; 269:451452.
74. Centers for Disease Control and Prevention. Malaria surveillanceUnited States,
1994. MMWR 1997; 46(SS-5):118.
75. Nahlen BL, Lobel HO, Cannon SE, Campbell CC. Reassessment of blood donor
selection criteria for United States travelers to malarious areas. Transfusion 1991;
31:798804.
76. Centers for Disease Control and Prevention. Addressing Emerging Infectious Disease
Threats. A Prevention Strategy for the United States. Atlanta: U.S. Department of
Health and Human Services, 1994.
19
Alternatives to Allogeneic Blood
and Strategies to Avoid Transfusion
Lawrence T. Goodnough
Washington University School of Medicine, St. Louis, Missouri
I. INTRODUCTION
Approximately 12 million red blood cell (RBC) units are transfused to nearly 4
million patients annually in the United States (1). The conservation of blood has
historically arisen from awareness that the inventory of this resource is limited
(2) as well as the knowledge that blood transfusion carries a risk (3). In addition,
emphasis on the costs of health care has raised issues related to the costs of
blood transfusion (46). Recent guidelines have emphasized that in the elective
transfusion setting, no blood transfusion is a desirable outcome (7). Further-
more, consensus conference recommendations (8) have emphasized that if blood
is to be transfused, autologous (the patients own) blood is preferable to al-
logeneic (from an anonymous, volunteer donor) blood. Thus the costs of blood
conservation, for which an increasing array of technological procedures and
products have become available, have also become an issue. The purpose of this
review is to provide an overview of emerging trends in blood transfusion and
blood conservation interventions in order to help identify areas important for
future investigation.
447
448 Goodnough
of anemia blood transfusion can reduce morbidity or mortality cannot be done for
ethical reasons. Attempts to reach conclusions from the considerable experience
obtained in managing Jehovahs Witnesses, who refuse blood on the basis of
religious beliefs, have been problematic (10). This is illustrated by the random-
ized prospective study of the synthetic oxygen carrier Fluosol-DA compared to
placebo in such a population, in which patients generally did well if their lowest
hematocrit (HCT) level exceeded 12% but did not do well at a HCT level of less
than 12%, irrespective of treatment group (11). One cannot reach conclusions
from these data regarding the transfusion trigger appropriate for populations
who would accept blood. Moreover, it is recognized that patients are heteroge-
neous, so that patients with medical (chronic, due to underproduction) anemias
are transfused differently from patients with surgical (acute, due to blood loss)
anemias (9). A more recent study (12) of surgical mortality in Jehovahs Witness
patients was able to show that preoperative hemoglobin levels and subsequent
blood loss were related to mortality, especially in patients known to have
cardiovascular disease. Attempts to link transfusions with outcomes such as
length of stay (13,14) have been unsuccessful. Two studies have found that
females are more likely to be transfused than are their male counterparts in the
elective surgical setting (15,16). The latter study found, even when using gener-
ous criteria to define excessive transfusions in which transfusion to replace losses
of less than 15% of the initial RBC volume was deemed inappropriate, that 21%
of patients undergoing joint replacement surgery received blood unnecessarily.
compared with the standard therapy group (5.9 3.8 vs. 9.8 9.4 units,
respectively). The improved patient care, along with reduced blood transfusions,
resulted in substantial economic savings. This approach has been described as a
powerful engine of change (31).
Approaches to red cell transfusion therapy in the surgical patient need to
acknowledge that patients are heterogeneous for risks related to anemia. In one
such approach to risk stratification for cardiac surgical patients has been pub-
lished (32), clinical variables determine whether a patient is considered at
standard or at increased risk for morbidity associated with cardiac surgery.
While complications related to anemia represent only one aspect of morbidity/
mortality in this setting, the physiological changes known to accompany acute
anemia (33) and the potential for myocardial tissue injury (34) suggest that risk
stratification for RBC transfusion decisions would be prudent. A recent analysis
of over 2000 cardiac bypass patients led to a model that calculated a transfusion
risk score, which was then validated prospectively in over 400 additional patients
(35). Now that cardiac surgery can be prospectively stratified, not only for
surgical risk but also for transfusion likelihood, the role of algorithms may be
especially productive in the standardization of blood transfusion and blood
conservation practices.
If transfusion outcomes can be predicted from patient-related factors, then
blood transfusion and blood conservation algorithms can be utilized to minimize
the variability of transfusion outcomes related to institutional (procedural) and
physician (transfusion practices) factors. Algorithms could take into account
patient heterogeneity by using the Higgins et al. (32) and McGovern et al. (35)
stratification of standard and increased risk patients. Blood transfusions and
blood-conservation strategies could be administered according to algorithms that
are incorporated into the daily practice of coronary revascularization (36). Such
an algorithm is illustrated in Figure 2. This approach can then enable physicians
to analyze transfusion outcomes, and the relationship of these outcomes, to
transfusion triggers, autologous blood procurement, pharmacological interven-
tions, and even emerging blood substitutes. The knowledge that we learn from
the standardization of these practices, and the comparison of transfusion out-
comes of different institutions or different therapeutic approaches, would form an
important database that could serve as a reference for the continuous improve-
ment of care in the surgical setting (37).
Figure 2 An algorithm approach for red cell transfusion in cardiac surgical patients
postoperatively. After establishing that the patients volume status is adequate, decisions
to transfuse would be based on hemoglobin/hematorcrit level, rate of blood loss, and
hemodynamic parameters [mean arterial pressure (MAP) and cardiac index (CI)]. Thresh-
olds for transfusion would differ for patients determined to be a low risk and high risk
for perisurgical complications. Mixed venous oxygen percent saturation (SVO2) could
serve as a physiological indicator of the balance between oxygen supply and demand for
transfusion decision making. (From Ref. 36.)
sion. This previously underutilized practice (38) has now become a standard of
care in elective surgical procedures such as orthopedic and urological surgery
(39,40). Guidelines on the criteria for utilization of this intervention, along with
selection of patients suitable for the technique, have been published (42,42).
Potential candidates for PAD prior to surgery include any patient scheduled for a
procedure for whom blood type and crossmatch is requested. Using this approach,
studies have found that, overall, 9% of autologous donors undergoing elective
surgery receive allogeneic blood (43). Under conditions of standard collection
(i.e., one unit weekly) the likelihood of exposure to allogeneic blood for autolo-
gous blood donors has ranged from 10% for radical prostatectomy (39) to 17%
for elective orthopedic surgery (44) to 56% for patients undergoing coronary
revascularization (45).
Blood Conservation Interventions 453
The efficacy of PAD is dependent on the degree to which the patients bone
marrow is stimulated to increase the production of red blood cells in order to
replace blood donated. Several studies have shown that the endogenous erythro-
poietin response is suboptimal at the level of mild anemia achieved during
collection of autologous blood (46,47). Under standard conditions of one
autologous unit donated weekly, a computer model demonstrated that if the
erythropoietic response to autologous blood phlebotomy is not able to maintain
hematocrit level during the donation interval, the predeposit of autologous blood
may actually be harmful (48).
In contrast to autologous blood donation under standard conditions,
studies of aggressive autologous blood phlebotomy (twice weekly for 3 weeks,
beginning 2535 days before surgery) have demonstrated that endogenous eryth-
ropoietin levels do rise when orthopedic surgical patients undergo a blood
donation (blood loss) of up to 1000 mL weekly during PAD. In a controlled
clinical trial of aggressive phlebotomy (49), serial erythropoietin levels showed a
logarithmic rise even in the Hgb concentration range of 110114 g/dL. This was
accompanied by significant erythropoiesis, in which a RBC volume equivalent to
three allogeneic blood units [at 200 mL each (50)] was generated over a
preoperative interval of 28 days (51). The erythropoietin response to blood loss
and the subsequent erythropoietic response have been shown to be independent
of patient gender or age (52). The ability of recombinant human erythropoietin
(EPO) to further accelerate the erythropoietic recovery from blood loss during
PAD has been demonstrated in several controlled trials (51,53,54). When aggres-
sive autologous phlebotomy was accompanied by simultaneous EPO administra-
tion, the equivalent of an additional two blood units (nearly five total) was
generated for subsequent blood conservation. Subsequent clinical trials have
demonstrated that EPO therapy during autologous blood donation can reduce the
need for allogeneic blood in patients undergoing elective orthopedic surgery
(55,56), even in patients with anemia of chronic inflammatory disease such as
rheumatoid arthritis (57).
Guidelines for autologous blood transfusion are controversial. Some pub-
lished guidelines recommend that the same criteria be used for utilization review
for autologous as are used for allogeneic blood units (57). This view is supported
by the risks of an immediate transfusion reaction, in which administrative error
and bacterial contamination account for nearly 50% of fatalities from transfusions
(58). These risks can occur for autologous blood units as well as for allogeneic
blood units. The alternative view holds that the risk/benefit relationship is lower
for autologous blood than for allogeneic blood, since the risk for disease trans-
missible by autologous blood has been reduced; this viewpoint argues for
different standards for transfusion of autologous than for allogeneic blood. While
transfusion services may choose to establish different guidelines for transfusion
454 Goodnough
particularly when blood is aspirated at vacuum settings greater than 100 torr.
The clinical importance of free hemoglobin in the concentrations usually seen has
not been established, although excess free hemoglobin may indicate inadequate
washing. Dilutional coagulopathy may occur if large volumes of salvaged blood
are administered (69).
As with preoperative autologous blood donation and acute preoperative
hemodilution, intraoperative autologous blood recovery should undergo scrutiny
concerning both safety and cost-effectiveness. A controlled study recently dem-
onstrated a lack of efficacy for intraoperative blood recovery when transfusion
requirements and clinical outcome were followed (70). A second study found that
only a minority of patients undergoing major orthopedic and cardiac surgery
achieved cost equivalence with intraoperative recovery using semi-automated
instruments compared with banked blood (71). While the recovery of a minimum
of one blood unit equivalent is possible for less expensive methods (with
unwashed blood), it is generally agreed that at least two blood unit equivalents
would need to be recovered using an automated recovery device (with washed
blood) in order to achieve cost-effectiveness (72). A recent study of this approach
in patients undergoing aortic aneurism repair concluded that this strategy is
particularly valuable in patients with at least 1000 mL surgical blood loss (73).
VI. CONCLUSION
The procurement of autologous blood has become a standard of care for elective
surgery in the United States. However, the costs and potential complications, as
Blood Conservation Interventions 457
REFERENCES
1. Wallace EL, et al. Collection and transfusion of blood in the United States, 1992.
Transfusion 1995; 35:802812.
2. Roche JK, Stengle JM. Open heart surgery and the demand for blood. JAMA 1973;
225:15161521.
3. Goodnough LT, Shuck JM. Review of risks, options, and informed consent for blood
transfusion in elective surgery. Am J Surg 1990; 159:602609.
4. Schwartz WB, Mendelson DN, Hospital cost containment in the 1980s. N Engl J Med
1991; 324:10371042.
5. Forbes JM, Anderson MD, Anderson GF, et al. Blood transfusion costs: a multicenter
study. Transfusion 1991; 31:319323.
6. Goodnough LT, Bodner MS, Martin JW. Blood transfusion and blood conservation:
cost and utilization issues. Amer J Med Qual 1994; 9:172183.
7. American College of Physicians. Strategies for elective red cell transfusion. Ann
Intern Med 1992; 116:403406.
8. National Institute of Health Consensus Conference. Perioperative red cell transfusion.
JAMA 1988; 260:27002703.
9. Welch GH, Meehan K, Goodnough LT. Prudent strategies for elective red cell
transfusion. Ann Intern Med 1992; 116:393403.
10. Mann MC, Votto J, Kambe J, McNamee MJ. Management of the severely anemic
patients who refuses transfusion: lessons learned during the care of a Jehovahs
witness. Ann Int Med 1992; 117:10421048.
11. Gould SA, Rosen AL, Sehgal LR, et al. Fluosol-DA As a red cell substitute in acute
anemia. N Engl J Med 1986; 314:19531956.
12. Carson JL, Duff A, Poses RM, et al. Effect of anamia and cardiovascular disease on
surgical mortality and morbidity. Lancet 1996; 348:10551060.
13. Kim DM, Brecher ME, Estes TJ, Morrey BF. Relationship of hemoglobin and
duration of hospitalization after total hip arthroplasty: implications for the transfusion
target. Mayo Clin Proc 1993; 68:3741.
14. Goodnough LT, Vizmig K, Riddell J IV, Soegiarso RW. Discharge haematocrit as
458 Goodnough
clinical indicator for blood transfusion audit in surgery patients. Transfusion Med
1994; 4:3544.
15. Friedman BA, Burns TL, Schork MA. An analysis of blood transfusion of surgical
patients by sex: a quest for the transfusion trigger. Transfusion 1980; 20:179188.
16. Goodnough LT, Verbrugge D, Vizmrg K, Riddell J. Identifying elective orthopaedic
surgical patients with amounts of blood in excess of need; the transfusion trigger
revisited. Transfusion 1992; 32:648653.
17. Toy PTCY. Effectiveness of transfusion audits and practice guidelines. Arch Pathol
Lab 1994; 118:435437.
18. Ayoub MM, Clark JA. Reduction of fresh frozen plasma use with a simple education
program. Am Surg 1989; 55:563565.
19. Shanberge JN. Reduction of fresh-frozen plasma use through a daily survey and
education program. Transfusion 1987; 27:226227.
20. Barnette RE, Fish DJ, Eisenstaedt RS. Modification of fresh-frozen transfusion
practices through educational intervention. Transfusion 1990; 30:253257.
21. McCullough J, Steeper TA, Connelly DP, Jackson B, Huntington S, Scott EP. Platelet
utilization in a university hospital. JAMA 1988; 259:24142418.
22. Simpson MB. Prospective-concurrent audits and medical consultation for platelet
transfusions. Transfusion 1987; 27:192195.
23. Lichtiger B, Fisher HE, Huh YO. Screening of transfusion service requests by the
blood bank pathologist: impact on cost containment. Lab Med 1988; 19:228230.
24. Hoeltge GA, Brown JC, Herzig RH, et al. Computer-assisted audits of blood
component transfusion. Cleve Clin J Med 1989; 56:267269.
25. Renner SW, Howanitz JH, Fishkin BG. Toward meaningful blood usage review: com-
prehensive monitoring of physician practice. QRB Qual Rev Bull 1987; 13:7680.
26. Goodnough LT. Retrospective utilization reviewAre we just going through the
motions? Arch Pathol Lab Med 1996; 120:802.
27. Audet AM, Goodnough LT, Parvin CA. Evaluating the appropriateness of red blood
cell transfusions: the limitations of retrospective medical record reviews. Int J Qual
Health Care 1996; 8:4149.
28. Lam HT, Schweitzer SO, Petz L, et al. Are retrospective peer-review transfusion
monitoring systems effective in reducing red blood cell utilization. Arch Pathol Lab
Med 1996; 120:810816.
29. Despotis GJ, Grishaber JE, Goodnough LT. The effect of an intraoperative treatment
algorithm on physician transfusion behavior in cardiac surgery. Transfusion 1994;
34:290296.
30. Despotis GJ, Santoro SA, Spitznagel E, Kater KM, Cox JL, Barnes P, Lappas DL.
Prospective evaluation and clinical utility of on-site coagulation monitoring in cardiac
surgical patients. J Thorac Cardiovasc Surg 1994; 107:271279.
31. Reinersten JL. Algorithms, guidelines, and protocols: can they really improve what
we do? Transfusion 1994; 34:281228.
32. Higgins TL, Estafanous FG, Loop FD, Beck GJ, Blum JM, Paranand L. Stratification
of morbidity and mortality outcome by preoperative risk factors in coronary artery
bypass patients. JAMA 1992; 267:23442348.
Blood Conservation Interventions 459
69. Horst HM, Dlugos S, Fath JJ, et al. Coagulopathy and intraoperative blood salvage.
J Trauma 1992; 32(5):646653.
70. Bell K, Stott K, Sinclair CJ, Walker WS, Gillon J. A controlled trial of intraoperative
autologous transfusion in cardiothoracic surgery measuring effect on transfusion
requirements and clinical outcome. Transfusion Med 1992; 2:295300.
71. Solomon MD, Rutledge ML, Kane LE, Yawn HD: Cost comparison of intraoperative
autologous versus homologous transfusion. Transfusion 1988; 28:379382.
72. Bovill DF, Moulton CW, Jackson WS, Jensen JK, Barcellos RW. The efficacy of
intraoperative autologous transfusion in major orthopaedic surgery: a regression
analysis. Orthopedics 1986; 9:14031407.
73. Goodnough LT, Monk TG, Sicard G, et al. Intraoperative salvage in patients
undergoing elective abdominal aortic aneurism repair: an analysis of costs and
benefit. J Vasc Surg 1996; 24:213218.
74. Semkiew LB, Schurman OJ, Goodman SB, Woolson ST. Postoperative blood salvage
using the cell saver after total joint arthroplasty. J Bone J Surg 1989; 71A:823827.
75. Flynn JC, Price CT, Zink WP. The third step of total autologous blood transfusion in
scoliosis surgery. Spine 1991; 16:S328329.
76. Faris PM, Ritter MA, Keating EM, Varleri CR: Unwashed filtered shed blood
collected after knee and hip arthroplasties. J Bone J Surg 1991; 73A:11691177.
77. Martin JW, Whiteside LA, Milliano MT, Reedy ME. Postoperative blood retrieval
and transfusion in cementless total knee arthroplasty. J Arthroplasty 1992; 7:205210.
78. Ayers DC, Murray DG, Duerr DM. Blood salvage after total hip arthroplasty. J Bone
Oint Surg 1995; 77A:13471351.
79. Goodnough LT, Verbrugge D, Marcus RE. The relationship between hematocrit,
blood lost, and blood transfused in total knee replacement: implications for post-
operative blood salvage and reinfusion. Am J Knee Surg 1995; 8:8387.
80. Mylod AG, France MP, Muser DE, Parsons JR. Perioperative blood loss associated
with total knee arthroplasty: a comparison of procedures performed with and without
cementing. J Bone J Surg 1990; 72A:110112.
81. McCarthy JC, Turner RH, Reuten JJ, Valeri CR, Ragno GM, Korten KW. The effect
of cell washing on the quality of shed blood in major reconstructive surgery. Orthop
Trans 1992; 16:484.
82. Clements DH, Sculco TP, Burke SW, Mayer K, Levine DB. Salvage and reinfusion
of postoperative sanguineous wound drainage. J Bone J Surg 1992; 74A:646651.
83. Woda R, Tetzlaff JE. Upper airway oedema following autologous blood transfusion
from a wound drainage system. Can J Anesth 1992; 39:290292.
84. Blevins FT, Shaw B, Valeri RC, Kasser J, Hall J. Reinfusion of shed blood
after orthopaedic procedures in children and adolescents. J Bone J Surg 1993;
75A:363371.
85. Kruskall MS, Umlas J. Acquired immunodeficiency syndrome and directed blood
donations: a dilemma for American medicine. Arch Surg 1988; 123:2325.
86. Thaler M, Shamiss A, Orgad S, et al. The role of blood from HLA-homozygous
donors in fatal transfusion-associated graft-vs-host disease. N Engl J Med 1989;
321:2528.
462 Goodnough
87. Perkins HA, Samson S, Busch MP. How well has self-exclusion worked? Transfusion
1988; 28:601602.
88. Chambers LA, Kruskall MS, Leonard SS, Ellis AM. Directed donor programs may
adversely affect autologous donor participation. Transfusion 1988; 28:645.
89. Goodnough LT. Predeposit of designated blood does not protect against homologous
blood exposure in patients who predeposit autologous blood for elective surgery. Am
J Clin Pathol 1989; 92:484487.
90. Ali S, Goodnough LT. An analysis of a directed donor program in 1995. Blood 1995;
86:852A.
20
Leukoreduction
Anne B. McDonald
Beth Israel Deaconess Hospital, Boston, Massachusetts
Walter H. Dzik
Beth Israel Deaconess Hospital and Harvard Medical School, Boston,
Massachusetts
I. INTRODUCTION
463
464 McDonald and Dzik
practiced. The process of centrifugation separates the least dense leukocytes from
the packed red cells, with more efficient removal of lymphocytes and monocytes
than granulocytes (1). In a variation of this processthe top and bottom
methodwhole blood is centrifuged at relatively high centrifugal force to com-
press both platelets and leukocytes into the buffy coat. The supernatant plasma is
then expressed out of the top of the primary bag into one satellite bag, while the
red cells are removed via the bottom into another. The buffy coat, composed of
donor white cells and platelets, is then pooled with further buffy coats and
centrifuged again at a lower centrifugal force. This allows separation of the
platelet-rich plasma from the white cell component, which is then discarded.
This method removes 7080% of leukocytes, a reduction sufficient to prevent
many febrile nonhemolytic transfusion reactions, but not other complications
whose prevention requires a higher degree of leukoreduction.
A variation of this method involves manipulation of the temperature of
whole blood prior to centrifugation. It was found that if the whole blood was first
cooled and then warmed prior to centrifugation, the removal of leukocytes could
be increased to 90%, with no apparent evidence of damage to red cells or
coagulation factors (2).
Other methods used in the past include washing and use of frozen/thawed
red cells. Washing is not the most efficient way to remove leukocytes because of
the wide variation in the number removed (3). Significant red cell loss may occur.
Because preparation takes place in an open system, washed red cells must
be used within 24 hours. Frozen deglycerolized red cells reduce the risk of
cytomegalovirus (CMV) transmission (4), but, because of the presence of viable
lymphocytes (5), they are inadequate to prevent graft-versus-host disease in
immunodeficient recipients.
Leukoreduction 465
B. Filtration
Advances in filtration technology have made consistent production of leuko-
reduced components a viable option. Table 2 summarizes the changes in filtration
over recent years.
1. Mechanism of Filtration
Removal of leukocytes by filtration results primarily from retention by a barrier.
The interior of most modern leukocyte removal filters consists of layers of
synthetic mesh made of nonwoven fibers. The design is such that the blood
entering the filter encounters a large surface area of the medium. Optimally, the
blood cannot bypass the filtration medium and the amount of blood retained in
the filter is minimal.
Adsorption of leukocytes to the synthetic fibers also contributes to leuko-
reduction. Modification of synthetic surfaces, by increasing wettability to
decrease the surface tension or by alteration in surface charge of the fibers (6),
has improved the performance of filters.
Biological mechanisms also influence leukoreduction. Electron micro-
scopic analysis of filters demonstrates that platelets undergo activation and
spreading on the surface of filter fibers during filtration of red blood cells.
Leukocytes, especially polymorphonuclear leukocytes, adhere to the activated
platelets and are removed (7). Platelets may also contribute by forming micro-
aggregates that trap cells (8).
be controlled to improve the quality of the end product. These factors are
summarized in Table 3.
The capacity of the filter used is of prime importance in the degree of
leukoreduction achieved. Current high-performance filters can reduce the residual
WBC content by 34 logs. The input load of the WBCs to be filtered has a direct
relation to the postfiltration WBC content. Manufacturers guidelines provide
filter capacity, or number of units of RBCs or platelets, for the average WBC load.
The cellular composition of the input product will affect the filtration process in
other ways, too. For example, elevated hematocrit in red cell concentrate may
result in slightly decreased leukocyte removal performance because of inhibited
leukocyte approach to adsorption points (9). The nature of the erythrocyte may
also affect filter performance. Filtration of hemoglobin AS (sickle cell trait) blood
results in poorer WBC removal than does filtration of hemoglobin AA blood
(10,11). It is thought that sickling of the red cells within the filter results in
obstruction of flow, preventing adequate contact of the leukocytes with the
filtration medium. Platelets in red cell concentrates also influence filtration.
Filters have decreased retention for fresh RBCs in the presence of platelets; this
has been attributed to biological interaction between platelets and WBCs that
promotes retention of the latter (8).
The flow rate through the filter is also a factor in leukoreduction; acceler-
ated blood flow (100 mL/min) is associated with decreased reduction (9). This
may result from partial detachment of adsorbed leukocytes due to accelerated
shear stress or from decreased contact time with the filtration medium. Alterna-
tively, slow filtration rate may result in insufficient pressure to ensure adequate
contact of leukocytes with the filtering medium or may allow an increase in
temperature resulting in increased leukocyte deformability (12). Studies have
shown that the efficiency of filtration is improved at refrigerated temperatures
(13). The reason for this may relate to decreased plasticity of leukocytes at lower
temperatures. The medium in which cells are suspended also influences the
efficiency of leukoreduction. The addition of small volumes of plasma to RBCs
3. Timing of Filtration
Leukocyte reduction can be performed before storage, before issuance from the
blood bank, or at the bedside (Table 4) (15). Prestorage leukoreduction is gaining
acceptance as the use of leukoreduced components becomes more routine. In-line
Advantages
May prevent accumula- Red cell units filtered in Conveniencefilter
tion of cytokines of short time span?im- added to intravenous
donor origin during proved leukoreduction. line at time of filtration.
storage.
May decrease risk of bac- Smaller no. of staff per-
terial contamination. forming procedure
less variability.
Leukoreduced units Easier to control quality
readily available on compared with bedside
request. filtration.
Easier to control quality Record of special needs
compared with bedside of particular patients.
filtration.
Disadvantages
Transfusion of Additional labor cost in Filtration occurs slowly
leukoreduced compo- blood bank. (24 h)red cell unit
nents to patients with May not remove leuko- warmsmay lead to
no defined clinical cyte fragments that less efficient filtration.
indication. accumulate during More difficult to institute
storage. quality control
Does not remove cyto- measures.
kines that may Units which fail quality
accumulate during control have already
platelet storage. been transfused.
Cost of filter. More staff involved in
processmore diffi-
cult to institute
standard procedures.
Cost of filter.
468 McDonald and Dzik
filtration (a blood bag system with integrated filter) allows for separation of
components and filtration without the need for opening the system, thereby
avoiding contamination and bacterial overgrowth during subsequent storage.
Prestorage leukoreduction has theoretical advantages of standardization and easy
availability of filtered units. Prestorage leukoreduction may decrease the inci-
dence of febrile nonhemolytic transfusion reactions (FNHTRs) from platelet
transfusion that results from passive transfer of cytokines. In an animal model
(16), the incidence of platelet refractoriness was decreased by prestorage leuko-
reduction. This may have resulted from removal of leukocyte fragments that
may not be removed during poststorage filtration. The issue of the importance
of leukocyte fragments has not been resolved. Some studies in an animal
model indicate a greater degree of platelet refractoriness following transfusion of
plasma supernatant of blood leukoreduced after storage when compared with that
leukoreduced before storage (17). However, it has also been demonstrated
that leukoreduction had no effect on soluble class I human leukocyte antigen
(HLA) substance, and that deliberately prepared leukocyte fragments bearing
HLA antigens did not seem to stimulate an alloimmune response in vitro (18).
There have not yet been any clinical trials in humans to determine the effect
of prestorage versus poststorage leukoreduction on prevention of alloimmuniza-
tion. Therefore, the issue remains unresolved.
The choice of filtration in the laboratory prior to issuance by the blood bank
versus filtration at the bedside would appear to many to be a minor issue in
leukoreduction. However, recent investigations have confirmed that bedside
filtration may be less than optimal in many cases for a variety of reasons. Bedside
filtration carries the advantage that expensive filters are restricted to units
transfused to patients for whom leukoreduction is indicated. However, concerns
have arisen regarding the quality of leukoreduction during bedside filtration.
Ledent and Berlin (12) found a 78% failure rate (>5 106 WBC/unit) when
leukoreduction was performed under conditions mimicking those of bedside
filtration. The failure rate when performed as in a blood bank setting was less
than 1%. The major difference in conditions was flow rate, with the rate at the
bedside very much slower than that in the laboratory. Slow filtration allows time
for warming of the unit, perhaps allowing increased leukocyte deformability and
thus less efficient removal of leukocytes. Deliberate warming of blood to 27C
resulted in a 20-fold increase in the number of leukocytes passing through the
filter (19), adding further weight to the importance of temperature control.
Therefore, the ideal time span in which to filter a unit of red cells would appear
to be slow enough so as not to generate excess shearing forces on the white cells
trapped in the filter, yet fast enough to minimize any potential warming of the
unit. Filtration at a relatively fast flow rate (by gravity over 1015 minutes), rather
than the slow flow at the bedside (over 23 hours), would appear to be the
most appropriate.
Leukoreduction 469
Method of %
leukoreduction removal Advantages Disadvantages
can be seen has not yet been determined. As such, development of improved
counting methods has paralleled the development of improved leukoreduction.
This is important in order to evaluate new technology critically and to gain a
better understanding of clinical trials involving leukoreduced components.
Methods to count residual leukocytes have used light or fluorescent micros-
copy, radioimmunoassays, flow cytometry, volumetric capillary cytometry, and
the polymerase chain reaction (PCR) (25). The most widely used method involves
a large volume (50 L) hemocytometer (Nageotte chamber). The sample to be
counted is mixed with a red cell or platelet lysing agent and a nuclear stain,
allowed to settle undisturbed in the counting chamber, and then examined under
200 magnification. When viewed by a trained observer, the method is simple
and has been shown to be accurate to approximately 1 WBC per L (26).
By concentrating a larger volume prior to sampling, the lower limit of detection
of Nageotte-based methods may be reduced even further (27).
Flow cytometry, by which a larger volume of sample can be analyzed,
achieves a lower limit of accurate detection of 0.1 WBC per L. Most flow
cytometric methods stain leukocytes with a fluorescent nuclear stain and analyze
the emission of light. The major disadvantage of this method is the need for
expensive instrumentation.
Other methods that have not yet achieved widespread use include volumet-
ric capillary cytometry and PCR-based counting methods. PCR-based methods
have an acceptable lower limit of detection, but have disadvantages of labor
intensity, equipment cost, and sample contamination.
and platelet refractoriness compared with unmodified pooled platelets. This trial
provides the best clinical evidence to date that leukoreduction prevents HLA
alloimmunization in patients undergoing cytoreductive chemotherapy.
A major issue for consideration is that HLA alloimmunization may be only
a poor surrogate marker for bleeding complications due to platelet refractoriness.
Because HLA alloimmunization is only one cause of refractoriness to platelet
transfusions, its elimination may not necessarily prevent this problem. Further-
more, serious bleeding complications are infrequent even when unmodified
components are used, and it may be difficult to justify the widespread use of a
costly procedure, such as leukoreduction, for a potentially small gain. Analysis of
this issue continues.
V. CONCLUSION
Leukoreduction has become an important consideration for choosing appropriate
blood components for transfusion. The major disadvantages of leukoreduction are
additional cost and loss of cellular components intended for transfusion. The po-
tential advantages of adequately leukoreduced components have been outlined.
Emphasis on quality control is essential to ensure that leukoreduced components
confer maximal benefit.
REFERENCES
1. Nakajo S, Chiba S, Takahashi TA, Sekiguchi S. Comparison of a top and bottom
system with a conventional quadruple-bag system for blood component preparation
and storage. In: Sekiguchi S, ed. Clinical Applications of Leukocyte Depletion.
Oxford: Blackwell Scientific Publications, 1982:1830.
2. Takahashi TA, Nakajo S, Chiba S, Hoseda M, Sekiguchi S. Cooling and warming
method for preparation of 1-log leukocyte-removed red cell concentrates using the
top and bottom method. In: Hogman CF, ed. Leukocyte Depletion of Blood Compo-
nentsPresent Trends and the Future. Amsterdam: VU University Press, 1994:145
148.
3. Wenz B. Clinical and laboratory precautions that reduce the adverse reactions,
allo-immunization, infectivity and possible immunomodulation associated with ho-
mologous transfusions. Trans Med Rev 1990; 4(suppl 1):37.
476 McDonald and Dzik
4. Tolkoff-Rubin NE, Rubin RH, Keller EE, et al. Cytomegalovirus infection in dialysis
patients and personnel. Ann Intern Med 1978; 89:625628.
5. Kurtz SR, Van Deinse WH, Valeri CR. The immunocompetence of residual leuko-
cytes at various stages of red cell cryopreservation with 40% w/v glycerol in an ionic
medium at 80C. Transfusion 1978; 18:441447.
6. Nishimura T, Kuroda T, Mizoguchi Y, et al. Advanced methods for leukocyte removal
by blood filtration. In: Brozovic B, ed. The Role of Leukocyte Depletion in Blood
Transfusion Practice. Oxford: Blackwell Scientific, 1989:3540.
7. Steneker I, van Luyn MJA, van Wachen PB, et al. Electron microscope examination
of white cell depletion on four leukocyte depletion filters. Transfusion 1992; 32:450
457.
8. Steneker I, Prins HK, Florie M, Loos JA, Biewenga J. Mechanism of leukocyte
depletion of red cell concentrates by filtration. The effect of the cellular composition
of the red cell concentrate. Transfusion 1993; 33:4250.
9. Nishimura T, Oka S, Yamawaki N. Technical aspects of leukocyte depletion. In:
Hogman CF, ed. Leucocyte Depletion of Blood ComponentsPresent Trends and
the Future. Amsterdam: VU University Press, 1994:5158.
10. Mijovic V, Kruse A. Filtration of blood from donors with HbAS: an unexpected
problem. In: Brozovic B, ed. The Role of Leucocyte Depletion in Blood Transfusion
Practice. Oxford: Blackwell Scientific, 1989:4850.
11. Bodensteiner D. White cell reduction in blood from donors with sickle cell trait
(letter). Transfusion 1994; 34:84.
12. Ledent E, Berlin G. Inadequate white cell reduction by bedside filtration of red cell
concentrates. Transfusion 1994; 34:765768.
13. Steneker I, Pieterz RNI, Reesink HW. Leukocyte depletion capacity in relation to
filtration temperature (abstr). Vox Sang 1994; 67(suppl 2):71.
14. Ledent E, Berlin G. Does plasma influence the efficiency of leukocyte filtration
(abstr)? Vox Sang 1994; 67(suppl 2):20.
15. Dzik WH. Leukoreduced components: you can filter now or you can filter later.
Transfus Sci 1992; 13:207210.
16. Engelfreit CP, Diepenhorst P, Gissen MVD, von Riesz E. Removal of leukocytes from
whole blood and erythrocyte suspensions by filtration through cotton wool. Vox Sang
1975; 28:8189.
17. Blajchman MA, Bardossy L, Carmen RA, Goldman M, Heddle NM, Singal DP. An
animal model of allogeneic donor platelet refractoriness: the effect of time of
leukoreduction. Blood 1992; 79:13711375.
18. Dzik S, Szulflad P, Eaves S. HLA antigens on leukocyte fragments and plasma
proteins: prestorage leukoreduction by filtration. Vox Sang 1994; 66:104111.
19. Sirchia G, Rebulla P, Sabbioneda L, Garcea F, Greppi N. Optimal conditions for white
cell reduction in red cells by filtration at the patients bedside. Transfusion 1996;
36:322327.
20. Sirchia G, Rebulla P, Parravicini A, Marangoni F, Cortelezzi A, Stefania A. Quality
control of red cell filtration at the patients bedside. Transfusion 1994; 34:2630.
Leukoreduction 477
21. Buchholz DH, AuBouchon JP, Snyder EL, et al. Removal of Yersinia enterocolitica
from AS-1 red cells. Transfusion 1992; 32:667672.
22. Hogman CF, Gong J, Eriksson L, Hambraeus A, Johansson CS. White cells protect
donor blood against bacterial contamination. Transfusion 1991; 31:612626.
23. Fitzpatrick JE, Orsini F, Pantano J. Residual leukocyte content of plateletpheresis
concentrates produced on the Haemonetics MCS+: evaluation of a method to predict
high post filter white cell counts (abstr). Transfusion 1995; 35(suppl 1):2S.
24. Riggert J, Zingsem J, Fourmel JJ, et al. European multicenter evaluation of the COBE
Spectra LRS leukoreduction system (abstr). Vox Sang 1996; 70(S2):11.
25. Dzik S. Principles of counting low numbers of leukocytes in leukoreduced blood
components. Trans Med Rev 1996;
26. Rebulla P, Dzik WH. Multicenter evaluation of methods for counting residual white
cells in leukocyte depleted red blood cells. Vox Sang 1994; 66:2532.
27. Sadoff BJ, Dooley DC, Kapoor V, Law P, Friedman LI, Stromberg RR. Methods for
counting a 6 log 10 white cell depletion in red cells. Transfusion 1991; 31:150156.
28. Dzik WH. Is the febrile response to transfusion due to donor or recipient cytokine
(letter)? Transfusion 1992; 32:594.
29. Heddle NM, Klama L, Singer J, et al. The role of plasma from platelet concentrates
in transfusion reactions. N Engl J Med 1994; 331:625628.
30. Goodnough LT, Riddell JV, Lazarus H, et al. Prevalence of platelet transfusion
reactions before and after implementation of leukocyte-depleted platelet concentrates
by filtration. Vox Sang 1993; 65:103107.
31. Aye MT, Palmer DS, Giulivi A, Hashemi S. Effect of filtration of platelet concentrates
on the accumulation of cytokines and platelet release factors during storage. Trans-
fusion 1995; 35:117124.
32. Heddle NM, Klama L, Kelton JG, Walker I, Meyer R, Levine M. Investigations of
two interventions to prevent acute reactions to platelets (abstr). Proceedings of the
24th congress of the ISBT, Mukuhari, Japan, 1996, p. 27.
33. Heddle NM. The efficiency of leukodepletion to improve platelet transfusion re-
sponse: a critical appraisal of clinical studies. Transfus Med Rev 1994; 8:1528.
34. Sintnicolaas K, van Marwijk KM, van Prooijen HC, et al. Leukocyte depletion of
random single-donor platelet transfusions does not prevent secondary human leuko-
cyte antigen-alloimmunization and refractoriness: a randomized prospective study.
Blood 1995; 85:824828.
35. Brady MT, Milam JD, Anderson DC, et al. Use of deglycerolized red blood cells to
prevent posttransfusion infection with cytomegalovirus in neonates. J Infect Dis
1984; 150:334339.
36. Bowden RA, Slichter SJ, Sayers M, Weisdorf D, Cays M, Schoch G, Banaji M, Haake
R, Welk K, Fisher L, McCullough J, Miller W. A comparison of filtered leukocyte-
reduced and cytomegalovirus (CMV) seronegative blood products for the preven-
tion of transfusion-associated CMV infection after marrow transplant. Blood 1995;
86:35993603.
37. van Prooijen HC, Visser JJ, van Oostendorp WR, de Gast GC, Verdonck LF.
Prevention of primary transfusion-associated cytomegalovirus infection in bone
478 McDonald and Dzik
marrow transplant recipients by the removal of white cells from blood components
with high-affinity filters. Br J Haematol 1994; 84:144147.
38. Busch MP, Lee TH, Heitman J. Allogeneic leukocytes but not therapeutic blood
elements induce reactivation and dissemination of latent human immunodeficiency
virus type 1 infection: implications for transfusion support of infected patients. Blood
1992; 80:21282135.
39. Dzik S, Blajchman MA, Blumberg N, Kirkley SA, Heal JM, Wood K. Current
research on the immunomodulatory effect of allogeneic blood transfusion. Vox Sang
1996; 70:187194.
21
Viral Inactivation
Bernard Horowitz
VITEX (V.I. Technologies, Inc.), New York, New York
Ehud Ben-Hur
Consultant in Photomedicine, New York, New York
I. INTRODUCTION
479
480 Horowitz and Ben-Hur
such as sugars, amino acids, and salts (7,8). These additives prevent unwanted
protein denaturation and loss of biological activity. The stabilization of the virus
by the added solute is much lower than that of the clotting factors (Table 1).
Treatment that achieves a sufficiently high level of virus elimination (over 6
log10) results in 6080% recovery of clotting factors. Nonenveloped viruses,
however, which tend to be heat-stable, are killed to a lesser extent by this process.
Other advantages of pasteurization are its ease of implementation in a factory
setting, avoidance of potentially toxic chemicals, and homogeneity of the treat-
ment (i.e., virus is inactivated at the same rate throughout the treated product).
B. Dry Heat
Virus inactivation by heating of lyophilized blood proteins occurs at higher
temperatures and takes a longer time than pasteurization because proteins are
stabilized in the absence of water. HIV can be eliminated from lyophilized Factor
VIII by heating at 68C for 96 hours (10). Recovery of Factor VIII activity can
be high under these conditions (11). However, because of heterogeneity of
lyophilized cakes with respect to solute and moisture content, heating has to be
conducted at 80C for 72 hours to achieve reproducible viral safety (12). Recov-
ery of Factor VIII activity exceeds 90%. As with pasteurization, noneveloped
viruses are more resistant to dry heat than are enveloped viruses, and their titer
(e.g., that of parvovirus) is reduced but not eliminated (13). A particular advantage
of dry heating is that it can be performed in the final container, eliminating the
possibility of posttreatment recontamination.
C. Solvent-Detergent
The use of the solvent tri(n-butyl)phosphate (TNBP), typically with Tween 80 or
other detergents, disrupts the viral lipid envelope with concomitant inactivation
of lipid-enveloped viruses (14). The treatment is rapid (46 h at 2430C), plasma
K (ln/hr)
proteins are not affected, and recovery of clotting factors can reach 100%.
The added solvent and detergent are removed after treatment, either simply in the
course of purifying the protein or with hydrophobic chromatography using a C18
resin. The safety of solvent-detergent (SD)treated blood products with respect
to HBV, HCV, and HIV is supported by studies in chimpanzees, 14 independent
clinical trials, and the preparation of HIVIG, a hyperimmune gamma globulin to
HIV prepared from HIV-infected donors (1517). A summary of viral safety from
clinical trials conducted with products treated by these methods is given in
Table 2. Advantages of SD treatment include its ease of implementation in a
factory setting and its very high level of virucidal action under conditions where
virtually all proteins are unaffected.
The excellent safety record of coagulation factors treated with solvent-
detergent encouraged the development of SD-treated plasma as a substitute for
fresh frozen plasma (FFP) (18). The procedure involves pooling of up to 2500
units of FFP, treatment with 1% TNBP and 1% Triton X-100 at 30C for 4 hours,
and removal of the reagents by hydrophobic chromatography. The final product
is sterile-filtered and frozen. Under these conditions, the rate of inactivation of
the model lipid-enveloped viruses, vesicular stomatitis virus (VSV) and Sindbis
virus, exceed those observed with Factor VIII concentrates. In addition, more than
106 infectious doses (ID50) of HBV, more than 105 ID50 of HCV, and more than
107.2 ID50 of HIV are killed. Approximately 15% of donor units contain anti-HAV
antibody. We have shown that more than 104.5 ID50 of HAV are neutralized in the
process (19). Because of pooling, SD-treated plasma has 30-fold more anti-HAV
antibody than intramuscular immune globulin, which is known to prevent HAV
spread, and has approximately the same quantity of antiparvovirus antibody as
intravenous immune globulin, a preparation used therapeutically to treat parvo-
2. Nanofiltration
Viruses can be removed by newly developed filters. The term nanofiltration
comes from the ability of the process to remove viruses as small as 15 nm (26,27).
The use of nanofiltration has the advantage of easy addition to existing processes.
484 Horowitz and Ben-Hur
4. Starch-Bound Iodine
Iodine is a strong oxidizing agent and, as a result, is a powerful microbicide.
However, in its free form iodine is not sufficiently selective. When bound to
polymers such as polyvinylpyrrolidone (34) and in particular crosslinked starch
(35), the virucidal action of iodine is more controlled. Thus, starch-bound iodine
at a concentration of 1.05 mg/mL resulted in more than 7 log10 inactivation of
model lipid enveloped and nonenveloped viruses, while more than 70% of
clotting factors activity in plasma was retained (35). Additional research is needed
to determine the efficacy of crosslinked starch-iodine on human pathogenic
viruses, as well as the effect on plasma proteins.
Viral Inactivation 485
B. Platelet Concentrates
The cellular components of blood are more difficult to sterilize than protein
solutions, because cell structure and function are disrupted more easily than
protein structure and function. In addition, infectious virus or its nucleic acid can
be harbored intracellularly. The challenge is eased somewhat because red blood
cells (RBCs) and platelets lack a nucleus and are nonreplicating. A decontaminat-
ing process must leave cellular function intact during both the treatment period
and subsequent storage. The storage period for platelets is 5 days, and the critical
functions require adhesion to subendothelial matrix proteins, aggregation, and
486 Horowitz and Ben-Hur
1. Methylene Blue
The absorption maximum of methylene blue (MB), 665 nm, is favorable for
sterilization of RBCs, and there is some clinical experience with its use for ster-
ilizing FFP (see above). However, there are problems associated with the use of
MB in RBCs. The main problem is the lack of inactivation by MB of cell-
associated HIV (40). Moreover, MB can photosensitize induction of HIV in
latently infected cells (60), and at a dose range in which sufficient virus elimina-
tion is achieved, the treatment causes RBC membrane damage (61). Other MB
derivatives are being studied that may circumvent these problems (62).
2. Benzoporphyrin Derivative
Benzoporphyrin derivative (BPD) is a photosensitizer undergoing clinical trials
for photodynamic treatment of skin cancer and other indications. BPD has an
absorption band at 692 nm and at concentrations of 24 g/mL plus 57 J/cm2 was
able to inactivate both cell-free and cell-associated HIV in whole blood (63).
There was only minimal hemolysis during 2 days of storage. Interestingly,
ziduvidine-resistant and -sensitive strains of HIV appear to be equally sensitive
to BPD photoinactivation. More work is required to evaluate RBC quality
following this virucidal treatment.
488 Horowitz and Ben-Hur
3. Hypericin
Initial work with hypericin suggested that this plant pigment may be an anti-HIV
agent (64). However, later studies indicate that most of the antiretroviral activity
of hypericin is light-dependent (65). This is in agreement with a large body of
literature on hypericin as a photodynamic agent. Maximal absorption of hypericin
occurs at 590 nm, and it is therefore not ideal for sterilization of RBCs. Even so,
there are efforts to optimize its virucidal potential by studies of structure-activity
relationships of several hypericin derivatives (66).
4. Phthalocyanines
Arguably, these are the most promising photosensitizers for sterilization of RBCs.
Phthalocyanines are porphyrin-like synthetic dyes. Because of their expanded
macrocycle, they absorb intensely at 660700 nm. Aluminum phthalocyanine
(AIPc) and its sulfonated derivatives are effective in photosensitizing inactivation
of lipid-enveloped viruses, including HIV (67). Other phthalocyanines were
shown to inactivate nonenveloped viruses (68). While tetrasulfonate AIPc caused
the least RBC damage under virucidal conditions, there was a need to eliminate
residual damage to erythrocytes, evidenced by reduced circulatory survival (69).
This was achieved by adding quenchers of ROS such as mannitol, glutathione,
and trolox (6971) prior to light exposure. While protecting RBCs, these quench-
ers had no effect on virus inactivation.
Other ways to reduce RBC damage while maintaining the virucidal potency
of phthalocyanines are the use of an appropriate light source to achieve high
irradiance (72) at a selective wavelength (73) and a special delivery vehicle (74).
These additional procedures to increase the specificity of the treatment were
required following the observation that inactivation of HIV in all its forms, as
well as inactivation of bloodborne parasites, was achieved only with the silicon
phthalocyanine Pc 4 (7577). Pc 4, however, caused more RBC damages in the
absence of these special precautions.
When RBCs are treated with Pc 4 and red light, taking into consideration
all of the above, virus sterilization can be achieved with little or no hemolysis
during storage (78). In vivo circulatory survival of rabbit RBCs is also close to
normal. Toxicological studies of Pc 4 are underway prior to evaluation of this
procedure in clinical trials.
5. Inactine
Inactine has recently been reported to inactivate both enveloped and nonenvel-
oped viruses. RBCs were reported to store well following treatment (79). The
structure of inactine has not bee disclosed, but it is said to covalently modify DNA
and to be mutagenic prior to but not following neutralization.
Viral Inactivation 489
V. CONCLUSIONS
The inactivation of viruses in blood proteins and plasma has made the transfusion
of these products absolutely safe with respect to transmission of HBV, HCV, and
HIV. The addition of nanofiltration, where applicable, or UVC irradiation to the
currently established methods (heat and SD treatment) should make these prod-
ucts safe also with respect to HAV and parvovirus B-19, the two nonenveloped
viruses reported to be transmitted by plasma derivatives. The remaining challenge
is the sterilization of red cell and platelet concentrates. The use of photosensitizers
for this purpose appears to be promising. PUVA is in clinical trials for platelets,
and Pc 4 is about to enter clinical trials for red cells. Table 3 summarizes the
Blood
Approach component Advantages Disadvantages
ACKNOWLEDGMENT
This work was supported in part by grant No. 2 RO1-HL 412221 from the
National Heart, Lung and Blood Institute.
REFERENCES
1. Schreiber GB, Busch MP, Kleinman SH, Korelitz JJ. The risk of transfusion-
transmitted viral infections. N Engl J Med 1996; 334:16851690.
2. Goedert JJ, Kessler CM, Aldort LM, Biggar RJ, Andes WA, White GC, Drummond
JE, Vaidya K, Mann DL, Eyster ME, Ragni MV, Lederman MM, Cohen AR, Bray
GL, Rosenberg PS, Friedman RM, Hilgartner MW, Blattner WA, Kroner B, Gail MH.
A prospective study of human immunodeficiency infection and the development of
AIDS in subjects with hemophilia. N Engl J Med 1989; 321:11411148.
3. Brettler DB, Alter HJ, Dienstag JL, Frosberg AD, Levine PH. The prevalence of
antibody to hepatitis C in a cohort of hemophilic patients. Blood 1990; 76:254.
4. Troisi CL, Hollinger FB, Hoots WK, Contant C, Gill J, Ragni M, Parmley R, Sexauer
C, Gomperts E, Buchanan G, Schwartz B, Adair S, Fields H. A multi-center study of
viral hepatitis in a United States hemophilic population. Blood 1993; 81:412428.
5. Oliphant JW, Hollander A. Homologous serum jaundice: experimental inactivation
of etiologic agent in serum by ultraviolet irradiation. Public Health Rep 1946;
61:598600.
6. Murray R, Oliphant JW, Tripp JT, Hampi B, Ratner F, Diefenbach WCL. Effect of
ultraviolet irradiation on the infectivity of ictrogenic plasma. J Am Med Assoc 1955;
157:814.
7. Hilfenhaus J, Herrman A, Mauler R, Prince AM. Inactivation of the AIDS-causing
retrovirus and other human viruses in antihemophilic plasma protein preparations by
pasteurization. Vox Sang 1986; 50:208211.
8. Hilfenhaus J, Weidmann E. Pasteurization as an efficient method to inactivate blood
borne viruses in factor VIII concentrates. Arzneimittelforschung 1986; 36:621625.
9. Horowitz B, Weibe ME, Lippin A, Vandersand J, Stryker MH. Inactivation of viruses
in labile blood derivatives. II. Physical methods. Transfusion 1985; 25:523527.
10. Levy JA, Mitra GA, Wong MF, Mozen MM. Inactivation by wet and dry heat of
AIDS-associated retroviruses during factor VIII purification from plasma. Lancet
1985; 1:14561457.
11. Hollinger B, Dolana G, Thomas W, Gyorkey F. Reduction in risk of hepatitis
transmission by heat treatment of a factor VIII concentrate. J Infect Dis 1984;
150:250262.
Viral Inactivation 491
12. Winkelman L, Owen NE, Evans DR, Evans H, Haddan ME, Smith JK, Prince PJ,
Williams JD, Lane RS. Severely heated therapeutic factor VIII concentrate of high
specific activity. Vox Sang 1989; 57:97103.
13. Mannucci PM. Clinical evaluation of viral safety of coagulation factor VIII and IX
concentrates. Vox Sang 1993; 64:197203.
14. Horowitz B, Wiebe ME, Lippin A, Stryker MH. Inactivation of viruses in labile blood
derivatives. I. Disruption of lipid-enveloped viruses by tri(n-butyl) phosphate deter-
gent combinations. Transfusion 1985; 25:516522.
15. Horowitz B, Prince AM, Hamman J, Watklevicz C. Viral safety of solvent/detergent-
treated blood products. Blood Coagul Fibrinol 1994; 5(suppl):S21S28.
16. Prince AM, Horowitz B, Baker L, Shulman RW, Ralph H, Valinsky J, Cundell A,
Brotman B, Boehle W, Rey F, Piet M, Reesink H, Lelie N, Tersmette M, Miedema
F, Barbosa L, Nemo G, Nastala CL, Allan JS, Lee DR, Eichberg JW. Failure of human
immunodeficiency virus (HIV) immune globulin to protect against experimental
challenge with HIV. Proc Natl Acad Sci USA 1988; 85:69446948.
17. Horowitz B. Inactivation of viruses found with plasma proteins: In: Goldstein J, ed.
Biotechnology of Blood. Stoneham: Butterworth-Heinemann, 1991:417430.
18. Horowitz B, Bonomo R, Prince AM, Chin SN, Brotman B, Shulman RW. Solvent/
detergent-treated plasma: a virus inactivated substitute for fresh frozen plasma. Blood
1992; 79:826831.
19. Lemon SM, Murphy PC, Smith A, Zou J, Hamman J, Horowitz B. Removal/neutral-
ization of hepatitis A virus during manufacture of high purity, solvent/detergent
treated plasma. Blood 1992; 79:826831.
20. Piquet Y, Janvier G, Selosse P, Doutremepuich C, Journeau J, Nicolle G, Platel D,
Vezon G. Virus inactivation of fresh frozen plasma by a solvent-detergent procedure:
biological results. Vox Sang 1992; 63:251256.
21. Hellstern P, Sachse H, Schwinn H, Overfrank K. Manufacture and in vitro character-
ization of solvent/detergent-treated human plasma. Vox Sang 1992; 63:178185.
22. Solheim BG, Svennevig JL, Mohr B, et al. The use of Octasplas in patients
undergoing open heart surgery. In: Muller-Berghanns G, et al., eds. DIS: Pathogene-
sis, Diagnosis and Therapy of Disseminated Intravascular Fibrin Formation. Amster-
dam: Elsevier Science Publishers, 1993:253262.
23. Moake J, Chintagumpala M, Turner J, McPherson P, Nolasco L, Steuber C, Santiago-
Borrero P, Horowitz M, Pehta J. Solvent/detergent-treated plasma suppresses shear-
induced platelet aggregation and prevents episodes of thrombotic thrombocytopenic
purpura. Blood 1994; 84:490497.
24. Pehta JC. Clinical studies with solvent-detergent-treated products. Transfusion Med
Rev 1996; 10:303311.
25. Piszkeiwicz D, Sun CS, Tondreau SC. Inactivation and removal of human
immunodeficiency virus in monoclonal purified antihemophilic factor (human)
(Hemophil M). Thromb Res 1989; 55:627634.
26. Hamamoto Y, Harada S, Kobayashi S, Yamaguchi K, Iijima H, Manabe S, Tsurumi
T, Aizawa H, Yamamoto N. A novel method for removal of human immunodeficiency
virus: filtration with porous polymeric membranes. Vox Sang 1989; 56:230236.
27. Burnouf-Radosevich M, Appourchaux P, Huart JJ, Burnouf T. Nanofiltration, a new
492 Horowitz and Ben-Hur
specific virus elimination method applied to high purity factor IV and factor XI
concentrates. Vox Sang 1994; 67:132138.
28. Horowitz B, Zou J, Hamman J, Gottlieb P, Chin S, Marx G, Golyakhovsky V.
Elimination of small, heat and solvent/detergent (S/D)-resistant viruses in factor VIII
concentrate and fibrin sealant. Throm Haemost 1995; 73:1460.
29. Patrick MH, Rahn RO. Photochemistry of DNA and polynucleotides: photoproducts.
In: Wang SY, ed. Photochemistry and Photobiology of Nucleic Acids, Vol. 2. New
York: Academic Press, 1976:3595.
30. Rauth AM. The physical state of viral nucleic acid and the sensitivity of viruses to
ultraviolet light. Biophys J 1965; 5:257273.
31. Chin S, Williams B, Gottlieb P, Margolis-Nunno H, Ben-Hur E, Hamman J, Jin R,
Dubovi E, Horowitz B. Virucidal UVC treatment of plasma and factor VIII concen-
trate: protection of proteins by antioxidants. Blood 1995; 86:43314336.
32. Marx G, Mou X, Freed R, Ben-Hur E, Yang C, Horowitz B. Protecting fibrinogen
with rutin during UVC irradiation for viral inactivation. Photochem Photobiol 1996;
63:541546.
33. Chin S, Jin R, Wang XL, Hamman J, Marx G, Mou X, Andersson I, Lindquist LO,
Horowitz B. Virucidal treatment of blood protein products with UVC. Photochem
Photobiol 1997; 65:432435.
34. Highsmith FA, Caple M, Walthall B, Shanbron E, Drohan WN. Viral inactivation
of vesicular stomatitis virus in normal human serum by crosslinked polyvinyl-
pyrrolidone. J Infect Dis 1993; 167:10271033.
35. Highsmith FA, Xue H, Caple M, Walthall B, Drohan WN, Shanbrom E. Inactivation
of lipid-enveloped and non-lipid-enveloped model viruses in normal human plasma
by crosslinked starch-iodine. Transfusion 1994; 34:322327.
36. Wallis C, Melnick JL. Photodynamic inactivation of animal viruses: a review.
Photochem Photobiol 1965; 4:159170.
37. Specht K. The role of DNA damage in PM2 viral inactivation by methylene blue
photosensitization. Photochem Photobiol 1994; 59:506514.
38. Abe H, Wagner SJ. Analysis of viral DNA, protein and envelope damage after
methylene blue, phthalocyanine derivative or merocyanine 540 photosensitization.
Photochem Photobiol 1995; 61:402409.
39. Lambrecht B, Mohr H, Knuver-Hopf J, Schmitt H. Photoinactivation of viruses in
human fresh plasma by phenothiazine dyes in combination with visible light. Vox
Sang 1991; 60:207213.
40. Wagner SJ, Robinette D, Storry J, Chen XY, Shumaker J, Benade L. Differential
sensitivities of viruses in red cells suspensions to methylene blue photosensitization.
Transfusion 1994; 34:521526.
41. Mohr H, Lambrecht B, Selz A. Photodynamic virus inactivation of blood compo-
nents. Immunol Invest 1995; 24:7385.
42. Wieding JU, Neumeyer H. Erste Erfahrungen mit Methylenblau-virus inaktiviertem
Fresh-frozen-plasma: Ergebnisse einer klinischen und einer in vitro Studie. Infusion
Ther 1992; 19:8490.
43. Mohr H, Knuver-Hopf J, Lambrecht B, Scheidecker H, Schmitt H. No evidence for
neoantigens in human plasma after photochemical virus inactivation. Ann Hematol
1992; 65:224228.
Viral Inactivation 493
44. Zeiler T, Riess H, Wittman G, Hintz G, Zimmerman R, Muller C, Geuft HG, Huhn
D. The effect of methylene blue phototreatment on plasma proteins and in vitro
coagulation capability of single-donor fresh-frozen plasma. Transfusion 1994;
34:685689.
45. Wagner SJ, Cifone MA, Murli H, Dodd RY, Myhr B. Mammalian genotoxicity
assessment of methylene blue in plasma: implications for virus inactivation. Transfu-
sion 1995; 35:407413.
46. Murphy S, Rebulla P, Bertolini F, Holme S, Moroff G, Snyder E, Stromberg R. In
vitro assessment of the quality of stored platelet concentrates. Transfusion Med Rev
1994; 8:2936.
47. Ben-Hur E, Song PS. Photochemistry and photobiology of furocoumarins
(psoralens). Adv Radiat Biol 1984; 11:131171.
48. Kao KJ. Effects of leukocyte depletion and UVB irradiation on allogenicity of major
histocompatibility antigens in platelet concentrates: a comparative study. Blood 1992;
80:29312937.
49. Heddle NM, Klama L, Singer J, Richards C, Fedak P, Walker I, Kelton JG. The role
of plasma from platelet concentrates in transfusion reactions. N Engl J Med 1994;
331:625628.
50. Lehmann PV, Shumm G, Moon D, Hurtenbach U, Falcioni F, Muller S, Zagy ZA.
Acute lethal graft-versus-host reaction induced by major histocompatibility complex
class II reactive T helper cell clone. J Exp Med 1990; 171:14851496.
51. Margolis-Nunno H, Robinson R, Ben-Hur E, Horowitz B. Quencher enhanced
specificity of psoralens photosensitized viral inactivation in platelet concentrates.
Transfusion 1994; 34:802810.
52. Lin L, Cook DN, Wiesehahn GP, Alfonso R, Behrman B, Cimino GD, Corten L,
Damonte PB, Dikeman R, Dupuis K, Fang YM, Hanson CV, Hearst JE, Lin CY,
Londe HF, Metchette K, Nerio AT, Pu JT, Reames AA, Rheinschmidt M, Tessman J,
Isaacs ST, Wollowitz S, Corash L. Photochemical inactivation of viruses and bacteria
in platelet concentrates by use of a novel psoralen and long wavelength ultraviolet
light. Transfusion 1997; 37:423435.
53. Margolis-Nunno H, Robinson R, Ben-Hur E, Chin S, Orme T, Horowitz B. Elimina-
tion of potential mutagenicity in platelet concentrates that are virally inactivated with
psoralens and ultraviolet A light. Transfusion 1995; 35:855862.
54. Margolis-Nunno H, Robinson R, Horowitz B, Geacintov NE and Ben-Hur E.
Psoralen-mediated virus photoinactivation in platelet concentrates: enhanced speci-
ficity of virus kill in the absence of shorter UVA wavelengths. Photochem Photobiol
1995; 62:917922.
55. Gottlieb P, Marolis-Nunno H, Robinson R, Shen LG, Chimezie E, Horowitz B,
Ben-Hur E. Inactivation of Trypanosoma cruzi trypomastigote forms in blood
components with a psoralen and ultraviolet A light. Photochem Photobiol 1996;
63:562565.
56. Lin L, Londe H, Janda JM, Hanson CV, Corash L. Photochemical inactivation of
pathogenic bacteria in human platelet concentrates. Blood 1994; 83:26982706.
57. Benade L, Shumaker J, Xu Y, Shen X, Dodd RY. Inactivation of free and cell-
associated human immunodeficiency virus in platelet suspensions by aminomethyl
psoralen and ultraviolet light. Transfusion 1994; 34:680684.
494 Horowitz and Ben-Hur
irradiance on virus sterilization and photodynamic damage in red blood cells sensi-
tized by phthalocyanines. Photochem Photobiol 1995; 61:190195.
73. Ben-Hur E, Zuk MM, Kenney ME, Oleinick NL, Mulvihill J, Horowitz B. Action
spectra (660-700 nm) for virus inactivation and red cell damage photosensitized by
the silicon phthalocyanine Pc 4. Lasers Med Sci 1996; 11:221225.
74. Ben-Hur E, Zuk MM, Chin S, Banerjee D, Kenney ME, Horowitz B. Biodistribu-
tion and virus inactivation efficacy of the silicon phthalocyanine Pc 4 in red blood
cell concentrates as a function of delivery vehicle. Photochem Photobiol 1995;
62:575579.
75. Margolis-Nunno H, Ben-Hur E, Gottlieb P, Robinson R, Oetjen J, Horowitz B.
Inactivation of multiple forms of human immunodeficiency virus in red cell concen-
trates by phthalocyanine photosensitization. Transfusion 1996; 36:743750.
76. Gottlieb P, Shen LG, Chimezie E, Bhang S, Kenney ME, Horowitz B, Ben-Hur E.
Inactivation of Trypanosoma cruzi trypomastigote forms in blood components by
photodynamic treatment with phthalocyanines. Photochem Photobiol 1995; 62:869
874.
77. Lustigman S, Ben-Hur E. Photosensitized inactivation of Plasmodium falciparum in
human erythrocytes by phthalocyanines. Transfusion 1995; 36:562565.
78. Ben-Hur E, Oetjen J, Zuk MM, Chan WS, Horowitz B. Photochemical decontamina-
tion of red cell concentrates with silicon phthalocyanine (Pc 4) and red light.
Transfusion 1996; 36(suppl):64S.
79. Zhang Q-X, Edson C, Budowsky E, Purmal A. Inactine: a method for viral inactiva-
tion in red blood cell concentrate. Transfusion 1998; 38(suppl):75S.
22
The Role of Quality in Blood Safety
Lucia M. Berte
Quality Systems Consultant, Elmhurst, Illinois
David E. Nevalainen
Quality Consultant, Health Care, Baileys Harbor, Wisconsin
Assuring blood transfusion safety means having error-free processes that begin
with donor selection and continue through blood component administration.
High-quality blood products and safe patient outcomes are best assured when
errors are prevented along the entire range of blood banking activity. Two goals
are desirable: (a) reduce to zero the number of blood components made from
unsuitable donations that result in any finished, labeled blood product that could
be distributed for clinical use, and (b) get the right blood products in the right
quantity to the right patient in the right place at the right time (1). These two broad
collections of processes and subprocesses involve different groups of people in
different locations at different times. However, a logical approach to apply
error-prevention initiatives across the entire range of activities can have the
desired end results.
497
498 Berte and Nevalainen
210.1 Status of cGMP A: General provisions A: General provisions A: Whole blood A: General provisions
210.2 Applicability of B: Organization and B: Organization and B: Red blood cells B: Quality system requirements
cGMP personnel personnel
210.3 Definitions C: Buildings and C: Plant and facilities C: Platelets C: Design controls
facilities
D: Equipment D: Equipment D: Plasma D: Document and record
controls
E: Control of E: (Reserved) E: (Reserved) E: Purchasing controls
components . . .
The Role of Quality in Blood Safety
perspective, blood banks cannot afford not to practice TPC if we are true to our
objective of providing safe, efficacious blood transfusions.
The essential principles of TPC are summarized in these statements from
an FDA guidance document (5):
Quality, safety, and effectiveness are built into a product.
Quality cannot be inspected or tested into a product.
Each step in the process must be controlled to meet quality standards.
These three statements provide facilities with a roadmap for what to do to assure
quality blood products and patient outcomes but do not describe how to do it.
Facilities determine for themselves what resources and methods they need to
apply to accomplish the intent of GMP. The important features of TPC are shown
in Figure 1.
Figure 1 The elements of Total Process Control (TPC) form a circular flow of process
analysis, SOP development, training, and assessment for which records document all
necessary activities. (From Ref. 14.)
The Role of Quality in Blood Safety 501
To best explain how the elements of TPC work, examples will be used.
An example for blood processing is a new screening test for a transfusion-
transmitted disease to be performed on each unit of donated blood. The example
for clinical transfusion is the implementation of a new armband system to
specifically identify transfusion recipients and link them with their compatibility
testing specimens. The following sections briefly describe the elements of TPC
in relation to these two examples and provide references for readers to access
important how-to details.
A. Validation
Before a new test, a new computer system or a new process is implemented, or
if there has been a significant change in an existing test method, instrument,
software, or process, an activity known as validation must take place. Process
validation is a requirement of cGMP Parts 211 (2) and 820 (4) for manufactured
pharmaceuticals and medical devices. It establishes documented evidence that a
specific process will consistently produce a product meeting its predetermined
specifications and quality characteristics. The facility must prepare a written
validation protocol that specifies what procedures and tests are to be conducted
and the data to be collected during practice runs of the new process. Only when
the data demonstrate that the process functions reliably and invariably can
standard operating procedures (SOPs) be finalized, remaining personnel trained,
and actual operations begun.
Table 2 outlines the activities of the major phases of a validation protocol
that are briefly described in the following paragraphs. Some details may vary
between processes to be validated, but the major concepts remain unchanged.
Validation examples specific to blood banking have been published (6).
1. Installation Qualification
In the example of a new blood donation screening test, all equipment used must
undergo installation qualificationactivities that demonstrate and document that
the equipment is suitable for its intended purpose, has been installed properly, and
is functioning as intended. For an enzyme immunoassay screening test, equip-
ment to be qualified would include specimen pipettors and dilutors, incubators,
tray washers, and spectrophotometric readers. Draft procedures for equipment
calibration, maintenance, quality control, and troubleshooting are first derived
from the directions included in the manufacturers equipment manuals, to which
are then added facility-specific details. Respective forms are designed to capture
measurements, data, and observations for the ongoing quality control program.
The new armband in our clinical transfusion example does not require installation
502
Installation qualification (IQ) Operational qualification (OQ) Performance qualification (PQ) Revalidation
Identification of items requiring Evaluation of process capabilities Development of testing plan Development of protocol
calibration
Determination of calibration Coordination of multiple pro- Predetermination of process/ Reaffirmation of IQ
method and schedule for cesses and operations product specifications
each item
Identification of items requiring Consideration of process variables Performance of process by Reaffirmation of OQ
maintenance operations personnel
Determination of maintenance Quantification of process variables Comparison of process out- Review of performance
methods and schedules for comes to specifications for history, OR
each item acceptability
Development of operating Determination of acceptable oper- Training and documentation Performance of a process run
procedures, including ating limits and variations
adjustments
Development of troubleshoot- Development of process Implementation of process Comparison of outcomes
ing procedures procedures to specifications for
acceptability
Development of monitoring and Retraining, if needed
control procedures
Listing of equipment parts Recommendation for
improvements
Training and documentation
Source: Ref. 7.
Berte and Nevalainen
The Role of Quality in Blood Safety 503
qualification unless barcode readers and/or electronic label printers are part of the
new armband-generation process.
2. Operational Qualification
This second phase of a validation protocol consists of evaluating the capability
of the processin this case, that the new blood screening test performs ac-
ceptably according to the manufacturers procedure using the newly qualified
equipment. The effectiveness and reproducibility of the new test is vigorously
challenged with conditions simulating those expected to be encountered during
actual production. In our testing example, challenges would include both strongly
and weakly reactive test samples, borderline reactive samples, and previous
proficiency testing material. In our armbanding example, the process would be
tested to see that barcodes and labels are readable and that the identification
linkages are traceable throughout specimen collection, compatibility testing, and
transfusion. The challenges should be repeated often enough to assure that the
results are consistent and meaningful. The draft testing procedures and related
forms are then readied for the validation runs.
3. Process Qualification
This phase of the validation protocol requires that selected operations personnel
(a) perform the new process as it would be done during actual operations,
(b) capture data about how the process worked, and (c) compare data to prede-
termined acceptance criteria. For the donor blood test, this final phase should not
proceed until operations and quality assurance personnel are confident that the
validation will be successful because of the high cost of performing full test runs.
In the testing example, a preset number of test runs would be performed by staff
members according to procedures derived from the manufacturers package insert
and facility-specific activities that were qualified in the preceding operational
qualification phase. In the armband example, all patients in a specified loca-
tion only would receive the new armband for a predetermined time. Any prob-
lems in the new processes are documented and followed up. Results of the
validations are reviewed by quality assurance personnel who reach a formal
conclusion as to the acceptability of the new process. Acceptance includes all
appropriate reviews and signatures and the finalization of, training in, and
activation of all related calibration, maintenance, quality control, troubleshooting,
and operating procedures.
4. Revalidation
Revalidation of manufacturing processes should be performed periodically and
whenever there are changes in raw materials, equipment, or processes that could
504 Berte and Nevalainen
affect the quality of the process outcome. Revalidation should also be considered
when there are significant changes in the processs quality performance history
as determined by statistical process control measurements. Revalidation consists
of reaffirming installation qualification and operational qualifications and either
performance of a validation run or comprehensive review of the processs quality
history records. In our testing example, revalidation would be required if the
manufacturer should change any test equipment, the test methodology, significant
reagents, or other variables of the blood screening test that could potentially affect
the quality of the test results. In our armband example, revalidation would be
required if there were changes to barcoding software or symbology or to label-
printing equipment.
archiving of all the processs SOPs and related forms. SOP management includes
the use of an SOP for how to write SOPs and another for how to change them.
Many external inspection deficiency citations involve lack of, outdated, or
incomplete SOPs or personnel found not to be following existing SOPs. These
problems lead to personal variations that can and have been shown to cause errors
that reduce blood safety and compromise the benefit of transfusion.
E. Monitoring
After the new procedures are validated, personnel trained, and a start-up date
selected, TPC requires ongoing monitoring of process variables to assure that the
process remains in control. A number of monitoring activities should be ongoing.
1. Quality Control
Laboratories have a long history of using quality control (QC) methods to assure
the accuracy of test results. For every test method or piece of equipment, there
are general and specific regulations and accreditation requirements for labora-
tories for QC monitoring and requirements for follow-up action when the results
506 Berte and Nevalainen
are out of acceptable range. The types of QC commonly performed in blood banks
are shown in Table 3. QC schedules are to be established and all results
documented and reviewed for trends and patterns that could suggest out-of-con-
trol performance. In the donor testing example, QC monitoring would include
positive and negative controls with every test run, blank, and background controls
for spectrophotometric measuring accuracy, monitoring of all temperature-regu-
lated equipment, and periodic recalibrations of pipettors and dilutors, among
other activities. The armband system needs no QC.
Equipment
Item Testing
Table 3 Continued
Equipment
Item Testing
Components
Item Testing
Reagents
Item Testing
Flowchart To identify the actual path that a product or service Symbols to depict process steps and decisions
follows to identify problems that lead to deviations
and errors
Check sheet To gather data based on sample observations to begin to Plot of defects vs. dates or times
detect patterns
Pareto chart To display relative importance of all problems or condi- Vertical bar graph with bars in descending
tions to a) identify basic cause of a problem, b) priori- order
tize problem solving, c) monitor success
Cause and effect diagram To identify, explore, and display the possible causes of a Fishbone
problem or condition
The Role of Quality in Blood Safety
Run chart To display trends with observation points over a speci- Plot of measurement vs. time
fied time period
Histogram To discover and display the distribution of data by bar Bar graph in frequency of distribution curve
graphing the number of amounts in each category
Scatter diagram To display what happens with one variable when another Plot of one variable on the x axis and the
variable changes to test a theory that the two variables other variable on the y axis
are related
Control chart To discover how much variability in a process is due to Run chart with statistically determined upper
random variation and how much is due to unique and lower control limit lines on either side
events to determine whether a process is in statistical of the process average
control
Process capability chart To determine whether the process, given its natural varia- Distribution curve showing allowable spread
tion, is capable of meeting established specifications of specification limits and measure of
actual process variation
Source: Ref. 8.
509
510 Berte and Nevalainen
F. Corrective Action
The monitoring activities of QC, SPC, occurrence reporting, and quality audit all
provide an overview of where the facilitys problems lie. The root cause of each
problem must be identified using a tool such as a cause-and-effect diagram (8)
and the problems categorized as system, knowledge, or behavior prior to taking
any corrective action. The chosen long-term corrective action must be appropriate
for the type of problem or the fix will not work. For example, training solutions
are often applied to system problems because it is easier to schedule a retraining
session than to dismantle a multistep process and rebuild it with error-prevention
steps to eliminate the problems root cause. In this scenario, follow-up monitoring
would, unfortunately, demonstrate that personnel have been further trained in a
process that still does not work.
G. Process Improvement
The time and resources expended in correcting quality problems are best utilized
when following a systematic approach for solving problems and improving
processes. The facility should adopt one of the common specialized problem
The Role of Quality in Blood Safety 511
solving models, such as those shown in Table 5, train personnel in its use, and
use the model for each performance improvement effort it undertakes. Most of
the improvement methods use multidisciplinary teams to work through the
successive steps.
Process improvements should not only be generated for problem areas.
When facilities truly practice continuous quality improvement, they will period-
ically reassess current processes to determine whether there may be a way to
perform them faster, better, or more cost-effectively. At any given time, there may
be several process-improvement efforts simultaneously underway as the facility
works through its prioritized list of problems and scheduled routine reviews.
Press, 1989.
dFrom Hospital Corporation of America, Nashville, TN.
eFrom Scholtes PR. The Team Handbook: How to Use Teams to
build quality into all processes and monitor the outcomes; current CFR cGMP
and laboratory regulations alone do not facilitate this coordination. To assist blood
banks in this effort, FDA introduced the Guideline for Quality Assurance in Blood
Establishments (11) as a draft in 1993; the document was finalized in 1995 after
considerable public input.
Concurrently with revision of the FDA draft guideline, the AABB produced
The Quality Program (12), a detailed expansion of the FDA guideline combining
the elements of TPC with the operational functions of blood banking from donor
selection through blood administration. Both FDA and AABB require blood
banks and transfusion services to have a quality program in place, though not
necessarily theirs. The AABB program, however, has the comprehension and
simplicity necessary for facilities of any size.
The framework of the AABB quality program is shown in Figure 2. The
elements of TPC are organized onto the left side of the quality program frame
and are collectively called the quality system. AABB refers to the TPC
elements as quality system essentials (QSEs). The important feature of the QSEs
is that they are to be uniformly applied to the operational functions shown across
the top of the frame. For example, training is to be given to all personnel in each
operational function and should minimally include organizational, safety, quality,
computer, and job-specific training. Likewise, the process for changing an SOP
should be the same in all the facilitys operational functions.
Whatever the size or scope of a blood bank facility (i.e., however many or
few activities or personnel in the operational functions at the top of the frame),
the QSEs remain unchanged because they are so fundamental to the facilitys
ability to build quality and safety into blood banking products and services.
To emphasize the importance of this concept, the AABB requires its institutional
members to have written quality policies that state what the facility intends to do
to implement the QSEs (TPC elements) in its operational functions. The quality
policies are supported by quality processes and procedures that describe how the
facility implements its stated intentions.
Figure 2 The AABB Quality Program grid showing cross-functional total process control elements (quality system essentials) applied to
513
A. Level A Documentation
As mentioned previously, the AABB requires that institutional members have a
written quality plan that states the facilitys quality objectives and policy. The
quality plan describes in a narrative or outline format what the facilitys policy is
with regard to each QSE (TPC element). The plan need not be lengthy, as other
documents will provide more detail for how the facility implements its quality
objectives. Sample quality plans are available to help facilities draft their own
(1315).
B. Level B Documents
Quality system processes describe the what, when, where, who of the TPC
elements/QSEs on the left side of the quality program frame. These documents
describe the processes for generating an SOP, training personnel, reporting an
occurrence, developing a validation protocol, and other tasks as shown in Table 6.
Quality system processes are cross-functionalthat is, they are applied across all
facility operating functions. For example, a person reporting an occurrence in the
component-processing area would follow the same procedure and use the same
reporting form as a person reporting an occurrence in the donor-testing area.
Important to note is that quality system processes often cross departmental lines
as to actions taken, responsibilities, and documentation to be generated and thus
have broad applicability. They are not written with the outline and detail of the
technical operating procedures used for the task specifics of employees jobs.
Flowcharts and tables are often used to describe processes.
C. Level C Documents
Level C documents are the technical operating procedures and related forms used
by employees in the facilitys operational functions. They are the work instruc-
tions that provide the detail necessary for personnel to perform their specific job
tasks. Using the donor testing example, a Level B quality process for SOP
development would be used to draft the Level C operating procedures for
calibration and maintenance of the spectrophotometric equipment, performance
of the immunoassay tests, reporting test results in the computer, and documenting
daily quality control. In the patient-identification example, the format described
in the same Level B SOP for SOPs would be used to write all the operating
procedures for patient specimen collection, compatibility testing, and blood
administration. Each Level C procedure may have one or more associated forms
that must be linked to it in some prescribed fashion as described in the Level B
process for the facilitys document control. All Level C procedures in one facility
The Role of Quality in Blood Safety 515
A. Self-Assessment
What should be assessed? The AABB has divided the scope of blood banking
activity into the operational functions shown across the top of the frame in Figure
2. For each operational function, processes and subprocesses have been identified.
For each process, one or more quality indicators have been identified that can be
used by facilities to monitor their performance over time (7). Operations person-
nel collect data and compare actual performance to preset expectations. Areas
needing improvement are prioritized, and the chosen continuous process improve-
ment model is applied. Follow-up monitoring of the quality indicators informs
the facility of its progress. Periodic reporting provides information for executive
management to make important decisions about resource allocations.
Self-assessment is an ongoing activity for which data should be collected
as part of the work process wherever possible. As an example from processing,
the log of unsuitable components should be updated as the components are
identified and quarantined. Occurrence reports should be generated as soon as
problems are discovered. Computers can be used to update blood donation,
deferral, and incomplete unit statistics as donor records are entered. As a
transfusion service example, persons performing compatibility testing can add to
the log of unacceptable specimens on specimen receipt in the laboratory. Com-
puters can be used to capture specimen receipt-to-result turnaround times for
emergent compatibility testing. Incomplete or incorrect transfusion report forms
can be logged on return to the transfusion service.
B. Quality Audits
Internal audits assess the conformance of the facilitys written quality system to
the standard and compare the actual activities to the written system. First-party
audits are performed by the facilitys own auditors as part of TPC monitoring.
Second-party audits are performed by facilities on their suppliers and take place
external to the facility. For example, a hospital blood bank may choose to perform
a second-party audit on the supplier of the blood components it procures for
transfusion. Third-party external quality audits are conducted by contracted
auditors to assess the facilitys quality system with respect to national or interna-
tional quality system standards and to provide registration or certification of
acceptable quality systems. The most common type of third-party audit is an ISO
9000 registration audit.
Table 7 is a comparison of the AABB QSEs to the ISO 9001 quality system
conformance model that demonstrates their compatibility. Some U.S. blood banks
have totally embraced the TPC and quality system philosophies and are preparing
for ISO 9000 registration audits. Two U.S. blood bank quality systems (16,17)
and several in other countries (18) have already become registered. Both the
518 Berte and Nevalainen
Table 7 Comparison of the AABB Quality System Essentials to the ISO 9001 Quality
System Conformance Model
AABB Quality System Essentials ISO 9001 Quality System Conformance Model
AABB and FDA have redesigned their inspection programs to better evaluate
whether the facilitys quality system conforms to regulations and accreditation
requirements and whether the facility has demonstrated compliance with and
effective implementation of its own quality policies, processes, and procedures.
VII. SUMMARY
The desirable and necessary patient outcome of safe, efficacious blood transfu-
sion requires a paradigm shift to the fervent belief that quality, safety, and
effectiveness must be built into all of a facilitys blood banking activities. In
manufacturing facilities, personnel are usually working in a single management
structure, which affords much more control in cooperation and coordination for
The Role of Quality in Blood Safety 519
REFERENCES
1. McClelland DBL, McMenamin JJ, Moores HM, Barbara JAJ. Reducing risks in
blood transfusion: process and outcome. Transfusion Med 1996; 6:110.
2. Food and Drug Administration, Department of Health and Human Services. 21 CFR
Parts 200-299. Washington, DC: U.S. Government Printing Office, revised annually.
3. Food and Drug Administration, Department of Health and Human Services. 21 CFR
Parts 600-799. Washington, DC: U.S. Government Printing Office, revised annually.
4. Food and Drug Administration, Department of Health and Human Services. 21 CFR
Part 820. Washington, DC: U.S. Government Printing Office, revised annually.
5. Food and Drug Administration, Department of Health and Human Services. Guide-
line on general principles of process validation. Washington, DC: U.S. Government
Printing Office, 1987.
6. Holliman S, ed. Validation in blood establishments and transfusion services.
Bethesda, MD: American Association of Blood Banks, 1996.
7. Nevalainen DE, Berte LM, Callery MF. Quality systems in the blood bank environ-
ment, 2d ed. Bethesda, MD: American Association of Blood Banks, 1998.
8. Brassard M, Ritter D. The memory jogger II. Methuen, MA: GOAL/QPC, 1994.
9. Standards for Blood Banks and Transfusion Services, 19th ed. Bethesda, MD:
American Association of Blood Banks, 1999.
10. Department of Health and Human Services. 42 CFR Parts 430 to end. Washington,
DC: U.S. Government Printing Office, revised annually.
11. Guideline on quality assurance in blood establishments; FDA Docket #91N-0450.
Bethesda, MD: U.S. Food and Drug Administration, 1995.
12. The quality program. Bethesda, MD: American Association of Blood Banks, 1994.
13. ISO Standards Compendium: ISO 9000 Quality Management, 6th ed. Geneva:
International Organization for Standardization, 1996.
14. Nevalainen DE, Callery MF. Module VIII: Quality system documentation. In: Quality
systems in the blood bank and laboratory environment. Bethesda, MD: American
Association of Blood Banks, 1994.
520 Berte and Nevalainen
15. AABB Transfusion Service Quality Assurance Committee. A model quality system
for the transfusion service. Bethesda, MD: American Association of Blood Banks,
1997.
16. South Texas Blood and Tissue receives ISO accreditation. AABB Weekly Report
1996; 2(44):4.
17. Denver blood center granted ISO 9000 certification. AABB Weekly Report 1998;
4(26):1.
18. Nevalainen DE, Lloyd HL. ISO 9000 quality standards: a model for blood banking?
Transfusion 1995; 35(6):521524.
23
Cost-Effectiveness Analysis of
Risk-Reduction Strategies
James P. AuBuchon
Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
I. INTRODUCTION
From the time of Hippocrates, the intention of doing no harm to ones patient has
been codified as a physicians duty. Viral transmission through transfusion clearly
represents an untoward outcome that both physician and patient would like to
avoid, and both parties would certainly prefer to minimize the risks of a
transfusion. Laudable and effective efforts have been directed at reducing trans-
fusion risks through donor screening, infectious disease marker testing, and
viral-inactivation efforts, but the risks of transfusion can only be lowered or
replaced, not eliminated. In addition to an ongoing balancing of risks and benefits,
the medical community is now also asked by society to achieve more with fewer
resources. The desire to reduce transfusion risk must be accomplished in an era
of finite resources. This situation is an appropriate one for application of decision
analysis toolsnot only to determine the relative return on investments in new
approaches to this health care problem, but also to illustrate where blood safety
efforts are most likely to yield substantial payoffs.
521
522 AuBuchon
wastage that is inevitable since the need for transfusion is so difficult to predict.
Through CEA, the magnitude of the benefits and costs have been quantitated for
specific situations and for general application (Table 1). The fact that PAD has
much poorer cost-effectiveness than most medical interventions (1921) in many
applications is not surprising given the relatively high level of safety of allogeneic
transfusion at present (15,22,23) and the need to collect many more units than are
necessary for transfusion in order to have autologous units available to cover the
potential need.
Through these analyses, however, those charged with delivering this option
to patients can gain valuable insights into how best to apply the technique. For
example, while it may be intuitive that collecting blood for a surgical procedure
that rarely results in transfusion is likely to be wasteful, the magnitude of the
impact of such use of PAD is clearly evident in the calculations of Etchason et al.,
who documented that cost-effectiveness varied exponentially with the likelihood
Cost-effectiveness
Intervention (cost/year of life extended)
Figure 1 Effects of patients age (15, 40, 60, or 70 years) and likelihood of the use of autologous units on
the cost-effectiveness of PAD. Note the exponential relationship between cost-effectiveness and proportion
of PAD units actually transfused. Patient age (i.e., projected longevity) also has a large impact on
cost-effectiveness calculations for PAD. Except in cases of long postoperative lifespan and high likelihood
AuBuchon
of use, PAD cost-effectiveness is far poorer than that of most medical and surgical interventions (which
usually have cost-effectiveness estimates better than $50,000/QALY). (Data adapted from Ref. 20.)
Cost-Effectiveness Analysis of Risk-Reduction Strategies 527
coronary artery disease so severe that surgery is necessary? While no study has
been of sufficient size to answer the question directly, a prospective donor also
needs to take into account that just delaying coronary revascularization in
order to allow for the collection of several units may have substantial mortality
riskapproximately 0.52% per month (29). These concerns call into question
whether PAD is the safest course of action for all patients.
Safety and efficacy questions extend to patients in more healthy states as
well. For example, 10% of patients donating for themselves before hysterectomy
ultimately needed to be transfused in a recent study, a transfusion rate 12 times
higher than that of patients who did not donate for themselves; the subset of
patients who donated preoperatively had lower admission hemoglobin values, and
autologous donation was an independent risk factor for transfusion (30). Obvi-
ously no benefit (at some cost and some, albeit small, risk) is achieved through
PAD when there is only a small likelihood of significant blood loss. Furthermore,
judicious use of erythropoetin to stimulate regeneration or expansion of red cell
mass and application of iron-replacement therapy may minimize complications
of repetitive phlebotomies (3133); however, any universal application of a
pharmacological adjunct or, indeed, any single PAD strategy cannot be supported.
Rather, judicious, patient-specific decisions must be made to identify the most
appropriate course of action based on the patients condition and the likelihood
of transfusion need, among other factors.
The cost-effectiveness analyses of PAD detailed above focused on preven-
tion of viral transmission as the primary benefit of using ones own blood for
surgery. Concerns have arisen over the last several years about the potential
immunosuppressive effect of allogeneic blood exposure and the possibility of
decreased tumor cell surveillance and increased risk of postoperative infection
following allogeneic transfusion (3437). Understanding the clinical significance
of these studies has been difficult because of the many confounding factors
embedded in the analyses and because of frankly contradictory results from
similarly designed studies. One group of researchers did include an estimate of
substantially increased risk of postoperative infection as a consequence of al-
logeneic transfusion in a CEA model of PAD before orthopedic surgery (38).
Inclusion of this complication led to the conclusion that PAD would be cost-
528 AuBuchon
effective or even cost-saving. On the other hand, a study performed for the
Canadian health care system concluded that even if the immunosuppressive
effects of allogeneic exposure were clinically significant, these would have a
relatively small impact on health care costs (39). Once the immunomodulatory
effects of allogeneic transfusion are well characterized and their clinical conse-
quences defined, any immunosuppressive effects of allogeneic transfusion could
be included in recalculation of a PAD model previously focused on viral trans-
mission. However, indications that leukoreduction may provide an alternative that
avoids this complication of allogeneic transfusion (40) raises another approach to
be considered: since a leukoreduction filter for a red cell unit generally costs
about half of the additional amount that a hospital must pay for an autologous
unit (41), leukoreduction may be a less costly and simpler approach to avoiding
this complication than PAD (42). For example, use of leukoreduced red cell units
in colorectal surgery resulted in a postoperative infection rate and length of
hospital stay similar to that of patients not requiring a transfusion and signifi-
cantly better than those receiving leukocyte-replete units. Additional studies to
define more precisely the relative costs and benefits of options to avoid compli-
cations such as this are necessary to clarify their cost-effectiveness.
may make salvage of red cells lost postoperatively not only feasible but also
cost-effective.
to donate for the purposes of discerning their HIV status rather than altruism.
Because no test is 100% sensitive (and some window period of negativity will
always exist following infection and infectivity), implementation of a new test to
make the blood supply safer could have its impact negated or ever reversed (60).
The greatest risk of this magnet effect is in situations where the incremental
sensitivity gained from implementation is small and the probability of infection
is relatively high. This combination (plus a high proportion of false positives that
would have erroneously been interpreted by donors as indicative of AIDS) led
blood bankers generally to avoid use of hepatitis B core antibody testing as a
surrogate marker for HIV infection prior to availability of HIV antibody testing
(61). The potential for the magnet effect to yield an unexpected, unfortunate, and
unintended result must be kept in mind when new interventions to improve blood
safety are being considered.
C. Microbial Inactivation
Additional increments in blood safety may be more readily achievable, however,
through microbial inactivation of blood components. These techniques would
have to be applied to all units, the vast majority of which were already free from
infectious agents, and thus cost-effectiveness would be expected to be rela-
tively poor unless the invervention were quite inexpensive. [For example, if
solvent-detergent (SD) treatment of frozen plasma (FP) cost $20 per unit, the
cost-effectiveness of this intervention would be close to $300,000/QALY (18).
Updating the cost-effectiveness to current infectious risks and prevailing costs
brings the cost-effectiveness of SD FP to approximately $210 million/QALY
(62,63).]
Beyond the obvious economic concern, the potential side effects of a
microbial inactivation treatment must also be considered. Any of the treatments
under consideration at present have the potential for leaving a small chemical
residual in the unit or generating a reactive intermediate molecule. This raises the
potential for chemical toxicity or mutagenicity following transfusion. Undoubt-
edly, toxicological tests will be performed prior to licensure documenting that
such untoward effects are exceedingly unlikely and/or small. However, when the
risk of transmission of a viral agent is also extremely small, just a minute
toxicological risk could outweigh all the benefit obtained from viral inactivation.
Manufacturers are clearly searching for ways to minimize residual agent follow-
ing inactivation, such as through adsorption filtration to remove methylene blue
from plasma (64), but this concern will have to be evaluated individually for each
proposed inactivation method.
The SD treatment process raises another issue of unintended negative
effects. For manufacturing logistics and economic reasons, this technique is
performed on pools of plasma rather than on individual units. If the technique
532 AuBuchon
D. Mistransfusion
Successful reduction of infectious disease risks through allogeneic transfusion
might have been predicted to lead to a redirection of attention to other transfusion
safety issues. Rather than this, however, additional effort has been directed toward
reducing these risks even further, ignoring estimates of low yield and concerns
about availability of financial support to match the desires for safer blood. As a
result, larger risks remain that might be addressed through interventions that are
more cost-effective than yet another infectious disease screening test.
An obvious example is mistransfusion and the risk of an ABO-related
fatality. The importance of ABO compatibility and the potentially catastrophic
Cost-Effectiveness Analysis of Risk-Reduction Strategies 533
E. Bacterial Contamination
The reappearance of concern regarding bacterial contamination (at least among
transfusion medicine professionals) highlights another dilemma faced by those
charged with providing a safe blood supply. While the risk of septic or endotoxic
shock after a red cell transfusion is on the order of 1 in 19,000,000 units, the
result is frequently fatal; far more platelet transfusions (perhaps 1/1,000113,000
platelet units) may harbor significant concentrations of bacteria, but because they
are more likely skin contaminants, their potential for inducing morbidity or
mortality is less (76). The source of these contaminants has been well described
534 AuBuchon
for the investment. These questions may be followed by a formal CEA, but other
factors also need to be considered.
Avoidance of HIV is a good example. Why is the public scared to death
of HIV transmission through transfusion yet apparently complacent about the
risks of mistransfusion?
A key factor may be the inevitability of premature death associated with
HIV infection. Surety of death as an outcome will prompt extreme measures
of avoidance (79,80). Although an intervention such as PAD will provide ac-
tual benefit to only a tiny minority of those using this transfusion option, all
patients donating blood for themselveseven those ultimately not needing a
transfusionreceive the psychological benefit of having taken steps to avoid a
dreaded outcome. Not only is the intensity of this drive significant, but it is poorly
captured in CEAs. As only tangible benefits are readily counted in the outcomes
(unless lingering psychic stress would alter quality of life or require medical
intervention), the importance of avoiding HIV for many people would not be fully
captured in an enumeration of the years of life extended by prevention of a certain
number of HIV transmissions.
Another facet of fear leads to a parallel problem. Activities in which
the untoward outcome is obvious and predictable are associated with less dread
than risks that occur in an unseen fashion and that occur indiscriminately,
that is, are not abetted by the actions of the victim. A transfusion-transmitted
infection would thus fall into the quadrant of highest dread since it occurs without
warning to an unsuspecting and undeserving victim and manifests only after a
period of latency (81). This situation, coupled with inordinate fear of HIV in
particular, as detailed above, may help explain the publics irrational fear of
HIV transmission via transfusion and the continued push for further reductions in
that risk.
Finally, the rule of rescue may apply to transfusion safety decisions (82).
In considering various therapeutic interventions, there is a human tendency to
favor implementation of approaches that result in preventing an imminent death
over those that would prolong the life of a patient beginning at some time in the
future. Although most blood safety initiatives are preventive rather than ther-
apeutic in nature, the concept of the rule of rescue may still be applicable since
the outcome of a blood safety initiative is often seen as preventing a premature
death. In addition, although measures taken to improve the safety of the blood
supply ultimately benefit only a small number of recipients, this group of patients
is readily identifiable and highly visible. In most situations, both lay and profes-
sional people will opt for the intervention that benefits a small, clearly identifiable
group of recipients over one that provides a similar or even greater aggregate
benefit spread in small increments over a large number of people (83,84). Thus,
the benefit of a blood safety initiative may have more psychological value than
as measured in QALYs.
536 AuBuchon
REFERENCES
1. McNeil BJ, Pauker SG, Sox HC, Jr, Tversky A. On the elicitation of preferences for
alternative therapies. N Engl J Med 1982; 306:12591262.
2. Nease RF, Jr. Risk attitudes in gambles involving length of life: aspirations, variations
and ruminations. Med Decis Making 1994; 14:201203.
3. Llewellyn-Thomas HA. Patients health-care decision making: a framework for
descriptive and experimental investigations. Med Decis Making 1995; 15:101105.
4. Sutherland HJ. Assessing patients preferences (editorial). Med Decis Making 1995;
15;286287.
5. Chapman GB, Elstein AS. Valuing the future: temporal discounting of health and
money. Med Decis Making 1995; 15:373386.
6. Beck JR, Pauker SG. The Markov process in medical prognosis. Med Decis Making
1983; 3:419458.
7. Russell LB, Gold MR, Siegel JE, Daniels N, Weinstein MC, for the Panel on
Cost-Effectiveness in Health and Medicine. The role of cost-effectiveness analysis in
health and medicine. JAMA 1996; 276:11721177.
8. Weinstein MC. Clinical Decision Analysis. Philadelphia: Saunders, 1980.
9. Eisenberg JM. Clinical economics. A guide to the economic analysis of clinical
practices. JAMA 1989; 262:28792886.
10. Weinstein MC, Stason WB. Foundation of cost-effectiveness analysis for health and
medical practices. NEJM 1977; 276:716721.
538 AuBuchon
11. Weinstein MC, Siegel JE, Gold MR, Kamlet MS, Russell LB, for the Panel on
Cost-Effectiveness in Health and Medicine. Recommendations of the Panel on
Cost-Effectiveness in Health and Medicine. JAMA 1996; 276:12531258.
12. Siegel JE, Weinstein MC, Russell LB, Gold MR, for the Panel on Cost-Effectiveness
in Health and Medicine. Recommendations for reporting cost-effectiveness analyses.
JAMA 1996; 276:13391341.
13. AuBuchon JP. Lessons learned from decision analysis. Transfusion 1996; 36:755
760.
14. AuBuchon JP. The role of decision analysis in transfusion medicine. Vox Sang 1996;
71:15.
15. Busch MP, Korelitz JJ, Kleinman SH, Lee SR, AuBuchon JP, Schreiber GB. Declin-
ing value of alanine aminotransferase in screening of blood donors to prevent
posttransfusion hepatitis B and C virus infection. The Retrovirus Epidemiology
Donor Stuedy. Transfusion 1995; 35:903910.
16. AuBuchon JP, Birkmeyer JD, Busch MP. Cost-effectiveness of expanded HIV test
protocols for donated blood. Transfusion 1997; 37:4551.
17. AuBuchon JP, Birkmeyer JD, Busch MP, Dodd RY, Lackritz EM, Petersen LR and
HIV Blood Donor Study Group. Cost-effectiveness of anti-HBc testing to reduce HIV
transmission risk (abstr). Transfusion 1996; 36:43S.
18. AuBuchon JP, Birkmeyer JD. Safety and cost-effectiveness of solvent-detergent
treated plasma. In search of a zero-risk blood supply. JAMA 1994; 272:12101214.
19. Birkmeyer JD, AuBuchon JP, Littenberg B, OConnor GT, Nease RF, Nugent WC,
Goodnough LT. The cost-effectiveness of preoperative autologous donation in coro-
nary bypass surgery. Ann Thoracic Surg 1994; 57:161169.
20. Birkmeyer JD, Goodnough LT, AuBuchon JP, Noordsij PG, Littenberg B. The
cost-effectiveness of preoperative autologous blood donation in total hip and knee
replacement. Transfusion 1993; 33:544551.
21. Etchason J, Petz L, Keeler E, Calhoun L, Kleinman S, Snider C, Fink A, Brook R.
The cost effectiveness of preoperative autologous blood donations. N Engl J Med
1995; 332:719724.
22. Lackritz EM, Satten GA, Aberle-Grasse J, Dodd RY, Raimondi VP, Janssen RS,
Lewis WF, Notari EP, IV, Petersen LR. Estimated risk of HIV transmission by
screened blood in the United States. N Engl J Med 1995; 333;17211725.
23. Schreiber GB, Busch MP, Kleinman SH, Korelitz JJ. The risk of transfusion-
transmitted viral infections. N Engl J Med 1996; 334:16851690.
24. Silvergleid AJ. All blood collected should be tested for infectious disease markers.
In: Maffei LM, Thurer RL, eds. Autologous blood transfusion: current issues.
Arlington, VA: American Association of Blood Banks, 1988:177182.
25. AuBuchon JP, Dodd RY. Analysis of the relative safety of autologous blood units
available for transfusion to homologous recipients. Transfusion 1988; 28:403405.
26. AuBuchon JP, Gettinger A, Littenberg B. Determinants of physician ordering of
preoperative autologous donations. Vox Sang 1994; 66:176181.
27. Popovsky MA, Whitaker B, Arnold NL. Severe outcomes of allogeneic and autolo-
gous blood donation: frequency and characterization. Transfusion 1995; 35:734737.
28. AuBuchon JP, Popovsky MA. The safety of preoperative autologous blood donation
in the non-hospital setting. Transfusion 1991; 31:513517.
Cost-Effectiveness Analysis of Risk-Reduction Strategies 539
29. Rankin JS, Hennein HA, Keith FM. The heart: I. Acquired diseases. In: Way LW, ed.
Current Surgical Diagnosis and Therapy. 10th ed. Norwalk, CT: Appleton & Lange,
1994:358382.
30. Kanter MH, van Maanen D, Anders KH, Castro F, Mya WW, Clark K. Preoperative
autologous blood donations before elective hysterectomy. JAMA 1996; 276:798801.
31. Brugnara C, Chambers LA, Malynn E, Goldberg MA, Kruskall MS. Red blood cell
regeneration induced by subcutaneous recombinant erythropoietin: iron-deficient
erythropoiesis in iron-replete subjects. Blood 1993; 81:956964.
32. Canadian Orthopedic Perioperative Erythropoietin Study Group. Effectiveness of
perioperative recombinant human erythropoietin in elective hip replacement. Lancet
1993; 341:12271232.
33. de Andrade JR, Jove M, Landon G, Frei D, Guilfoyle M, Young DC. Baseline
hemoglobin as a predictor of risk of transfusion and response to epoetin alfa in
othopedic surgery patients. Am J Orthop 1996; 25:533542.
34. Vamvakas EC, Taswell HF. Blood transfusion as a predictor of long-term mortality
(abstr). Transfusion 1993; 33:28S.
35. Vamvakas EC, Moore SB. Blood transfusion and postoperative septic complications.
Transfusion 1994; 34:714727.
36. Vamvakas E, Moore SB. Perioperative blood transfusion and colorectal cancer
recurrence: a qualitative statistical overview and meta-analysis. Transfusion 1993;
33:754765.
37. Vamvakas EC, Moore SB. Confounding and the effect of allogeneic transfusion on
survival (letter). Vox Sang 1995; 69:142143.
38. Healy HC, Frankforter SA, Graves BK, Reddy RL, Beck JR. Preoperative autologous
blood donation in total-hip arthroplasty. A cost-effectiveness analysis. Arch Pathol
Lab Med 1994; 118:465470.
39. Canadian Coordinating Office for Health Technology Assessment. Leukoreduction:
the techniques used, their effectiveness and costs. Ottawa: Canadian Coordinating
Office for Health Technology Assessment (CCOHTA), 1998.
40. Jensen LS, Kissmeyer-Nielsen P, Wolff B, Qvist N. Randomised comparison of
leucocyte-depleted versus buffy-coat-poor blood transfusion and complications after
colorectal surgery. Lancet 1996; 348:841845.
41. Forbes JM, Anderson MD, Anderson GF, Breecker GC, Rossi EC, Moss GS. Blood
transfusion costs: a multicenter study. Transfusion 1991; 31:318323.
42. Jensen LS, Grunnet N, Hanberg-Sorensen F, Jorgensen J. Cost-effectiveness of blood
transfusion and white cell reduction in elective colorectal surgery. Transfusion 1995;
35:719722.
43. Cohen JA, Brecher ME. Preoperative autologous blood donation: benefit or detri-
ment? A mathematical analysis. Transfusion 1995; 35:640644.
44. Goodnough LT, Monk TG, Brecher ME. Autologous blood procurement in the
surgical setting: Lessons learned in the last 10 years. Vox Sang 1996; 71:133141.
45. Monk TG, Goodnough LT, Birkmeyer JD, Brecher ME, Catalona WJ. Acute normo-
volemic hemodlution is a cost-effective alternative to preoperative autologous dona-
tion in patients undergoing radical retropubic prostatectomy. Transfusion 1995;
35:559565.
540 AuBuchon
46. AuBuchon JP. Blood transfusion options: improving outcomes and reducing costs.
Arch Pathol Lab Med 1997; 121:4047.
47. Brecher ME, Goodnough LT, Monk T. The value of oxygen-carrying solutions in
the operative setting, as determined by mathematical modeling. Transfusion 1999;
39:396402.
48. Solomon MD, Rutledge ML, Kane LE, Yawn DH. Cost comparison of intraoperative
autologous versus homologous transfusion. Transfusion 1988; 28:379382.
49. Popovsky MA, Devine PA, Taswell HF. Intraoperative autologous transfusion. Mayo
Clin Proc 1985; 60:125134.
50. AuBuchon JP. Autologous transfusion and directed donations: current controversies
and future directions. Transf Med Rev 1989; 3:290306.
51. Solomon MD, Rutledge MI, Kane LE, Yawn DH. Cost comparison of intraoperative
autologous versus homologous transfusion. Transfusion 1988; 28:379382.
52. Rumisek JD, Weddle RL. Autotransfusion in penetrating abdominal trauma. In: Hauer
JM, Thurer RL, Dawson RB, eds. Autotransfusion. New York: Elsevier/North Hol-
land, 1981:105113.
53. Karczewski DM, Lema MJ, Glaves D. The efficiency of an autotransfusion system
for tumor cell removal from blood salvaged during cancer surgery. Anesth Analg
1994; 78:11311135.
54. Umlas J, Foster RR, Dalal SA, OLeary SM, Garcia L, Kruskall MS. Red cell loss
following orthopedic surgery: the case against postoperative blood salvage. Transfu-
sion 1994; 34:402406.
55. Umlas J, Sager A. Cost-effectiveness of recovering blood post-arthroplasty (abstr.)
Transfusion 1994; 34:27S.
56. Jackson BR, Umlas J, AuBuchon JP. The cost-effectiveness of postoperative red cell
recovery for preventing transfusion-associated viral transmission following orthope-
dic surgery. Transfusion (in press).
57. Infectious disease testing for blood transfusions. NIH Consensus Development Panel
on Infectious Disease Testing for Blood Transfusions. JAMA 1995; 274:13741379.
58. AuBuchon JP, Birkmeyer JD, Busch MP. Blood Safety: Challenges and opportunities.
Ann Int Med 1997; 127:904909.
59. Leitman SF, Klein HG, Melpolder JJ, Read EJ, Estaban JI, Leonard EM, Harvath L,
Shih W-K, Nealon R, Foy R, Darr F, Alter HJ. Clinical implications of positive tests
for antibodies to human immunodeficiency virus type 1 in asymptomatic blood
donors. N Engl J Med 1989; 321:917924.
60. Korelitz JJ, Busch MP, Williams AE for the Retrovirus Epidemiology Donor Study.
Antigen testing for human immunodeficiency virus (HIV) and the magnet effect: Will
the benefit of a new HIV test be offset by the numbers of higher-risk, test-seeking
donors attracted to blood centers. Transfusion 1996; 36:203208.
61. Zuck TF, Eyster ME. Blood safety decisions, 1982-1986: perceptions and misconcep-
tions. Transfusion 1986; 36:928931.
62. Pereira A. Cost-effectiveness of transfusing virus-inactivated plasma instead of
standard plasma. Transfusion 1999; 39:479487.
Cost-Effectiveness Analysis of Risk-Reduction Strategies 541
63. Jackson BR, AuBuchon JP, Birkmeyer JD. Update of cost-effectiveness analysis for
solvent-detergent treated plasma. JAMA 1999; 282:329.
64. AuBuchon JP, Pickard C, Herschel L, OConnor JL, Williams S. Preparation of
methylene blue (MB) plasma with reduction in residual MB content by adsorption
filtration (abstr). Transfusion 1996; 36:65S.
65. Lynch TJ, Weinstein MJ, Tankersley DL, Fratantoni JC, Finalyson JS. Considerations
of pool size in the manufacture of plasma derivatives. Transfusion 1996; 36:770775.
66. Busch M, Glynn S, Schreiber G for the Retrovirus Epidemiology Donor Study.
Increased risk of viral transmission due to exclusion of older donors because of
Creutzfeld-Jakob disease (CJD) (abstr). Transfusion 1996; 36:59S.
67. Sazama K. Reports of 355 transfusion-associated deaths: 1976 through 1985. Trans-
fusion 1990; 30:583590.
68. Honig CL, Bove JR. Transfusion-associated fatalities: review of Bureau of Biologics
reports. Transfusion 1980; 20:653661.
69. Linden JV, Paul B, Dressler KP. A report of 104 transfusion errors in New York State.
Transfusion 1992; 32:601606.
70. Renner SW, Howanitz PJ, Bachner P. Wristband identification error reporting in 712
hospitals. A College of American Pathologists Q-Probes study of quality issues in
transfusion practice. Arch Pathol Lab Med 1993; 117:571577.
71. Reason J. Human Error. Cambridge: Cambridge University Press, 1990.
72. Voelker R. Treat systems, not errors, experts say. JAMA 1996; 276:15371538.
73. Baele PL, De Bruyere M, Deneys V, Dupont E, Flament J, Lambermont M, D,
Steensens L, van Camp B, Waterloos H. Bedside transfusion errors. A prospective
study by the Belgium SAnGUIS group. Vox Sang 1994; 66:117121.
74. Wenz B, Burns ER. Improvement in transfusion safety using a new blood unit and
patient identification system as part of safe transfusion practice. Transfusion 1991;
31:401403.
75. AuBuchon JP, Littenberg B. A cost-effectiveness analysis of using a mechanical
barrier system to reduce the risk of mistransfusion. Transfusion 1996; 36:222226.
76. Wagner SJ, Friedman LI, Dodd RY. Transfusion-associated bacterial sepsis. Clin
Microbiol Rev 1994; 7:290302.
77. Brecher ME, Hogan JJ, Boothe E, Kerr A, McClannan L, Jacobs MR, Yomtovian R,
Chongokolwatana V, Tegtmeier G, Henderson S, Pineda A, Halling V, Kemper M,
Kuramato K, Holland PV, Longiaru. Platelet bacterial contamination and the use of
a chemiluminescence-linked universal bacterial ribosomal RNA gene probe. Trans-
fusion 1994; 34:750755.
78. Vamvakas EC, Taswell HF. Long-term survival after blood transfusion. Transfusion
1994; 34;471477.
79. Fowler FJ, Jr, Cleary PD, Massagli MP, Weissman J, Epstein A. The role of reluctance
to give up life in the measurement of the values of health states. Med Decis Making
1995; 15:195200.
80. Schneiderman LJ, Kaplan RM. Fear of dying and HIV infection vs hepatitis B
infection. Am J Publ Health 1992; 82:584586.
81. Morgan MG. Risk analysis and management. Sci Am 1993; 269:3235, 3841.
542 AuBuchon
82. Hadorn DC. Setting health care priorities in Oregon. Cost-effectiveness meets the rule
of rescue. JAMA 1991; 265:22182225.
83. Hux JE, Leviton CM, Naylor CD. Prescribing propensity: influence of life-expectancy
gains and drug costs. J Gen Intern Med 1994; 9:195201.
84. Rose G. Sick individuals and sick populations. Int J Epidemiol 1985; 14:3238.
85. Bowden RA, Slichter SJ, Sayers M, Weisdorf D, Cays M, Schoch G, Banaji M, Haake
R, Welk K, Fisher L, McCullough J, Miller W. A comparison of filtered leukocyte-
reduced and cytomegalovirus (CMV) seronegative blood products for the preven-
tion of transfusion-associated CMV infection after marrow transplant. Blood 1995;
86:35993603.
24
Red Blood Cell Substitutes
I. INTRODUCTION
A. Background
During the last 15 years there has been a rapid development in the number of
materials with the ability to transport and deliver oxygen and other gases. While
many of them were conceived as substitutes for red blood cells (RBC),* it is
becoming apparent that their unique characteristics suit them for new applications
beyond the scope of RBC. In this chapter, the different approaches that are being
taken to develop non-RBC oxygen carriers will be discussed and the status of the
clinical trials in progress as of June 2000 will be reviewed.
*The terms blood substitute and RBC substitute are used commonly, including in medical
literature databases. The term RBC substitute will be used in this chapter recognizing that these
oxygen-carrying materials are not intended to replicate all of the functions of the many elements of
whole blood and that they have characteristics that suit them for clinical roles beyond those possible
for the erythrocyte.
543
544 Szczepiorkowski and Stowell
Characteristics Requirement
F F
c F c F
F c F c
F F
STRUCTURE
Red Blood Cell Substitutes
150 300 450 50 100 150 50 100 150 50 100 150 50 100 150
O2 SATURATION
CONCENTRATION
CONCENTRATION
CONCENTRATION
CONCENTRATION
CONCENTRATION
CLEARANCE
12 24 60 120 12 24 60 120 12 24 60 120 12 24 60 120 12 24 60 120
hours days hours days hours days hours days hours days
Figure 1 Comparison of several characteristics of three types of RBC substitutes to one another and to erythrocytes: structure,
oxygen binding, and intravascular clearance. The dotted lines in the panels for the RBC substitutes represent the behavior of
the erythrocyte and are shown for comparison. The RBC substitutes are perfluorocarbon (PFC), unmodified and modified
547
Advantages Disadvantages
Product
(manufacturer) Perfluorocarbon Trial level Application
aInformationcurrent to 6/00.
PCTA, Percutaneous, transluminal coronary angioplasty; CABG, coronary artery bypass graft; ANH,
acute normovolemic hemodilution; GI, gastrointestinal; IRDS, infant respiratory distress syndrome;
ARDS, adult respiratory distress syndrome.
2. Oxyfluor
HemaGen/PFC formulated a perfluorocarbon emulsion named Oxyfluor consist-
ing of perfluorodichlorooctane stabilized with lecithin and safflower oil, which
may be stored for a year at room temperature. Animal studies showed that
Oxyfluor was capable of delivering oxygen in models of shock resuscitation and
surgical bleeding and was well tolerated. Lung hyperinflation was noted in the
treated animals, as has been observed with many of the perflurocarbons, although
it has not emerged as a significant problem in humans. Safety studies in humans
have demonstrated that thrombocytopenia and a flu-like syndrome occur with this
perfluorocarbon preparation, as was the case with Oxygent.
Although Oxyfluor underwent phase II trials in surgery patients,
HemaGen/PFC chose to exploit the ability of perfluorocarbons to dissolve gases
by using Oxyfluor to remove the microbubbles that form in the circulation of
patients undergoing cardiopulmonary bypass. These microbubbles are thought to
embolize to the microvasculature of the brain, where they produce the neurolog-
ical and neuropsychiatric changes seen in some patients following bypass (13,14).
In animal cardiopulmonary bypass model systems, Oxyfluor was effective in
scavenging microbubbles and reducing the formation of these microemboli (15).
HemaGen/PFC ceased operations for financial reasons. Hence, Oxyfluor is no
longer in development.
Advantages Disadvantages
Product Hemoglobin
(manufacturer) source Trial level Application
introduced to correct the leftward shift that occurs when hemoglobin is removed
from the influence of 2,3-DPG.
Some of the advantages and disadvantages of the recombinant hemoglobins
are summarized in Table 7. While the opportunity for engineering recombinant
hemoglobin and the potential of tailoring it for different applications are exciting,
Somatogen has faced unprecedented challenges in scaling up the production and
purification processes. They have, however, reported success in operating a
50,000 L production-level fermenter.
A series of phase I and II trials were completed with this recombinant
hemoglobin preparation, Optro, which was generally well tolerated. Mild gastro-
intestinal symptoms were reported with this hemoglobin preparation as with
others (37).
One of the more novel properties of this product is an erythropoietic effect,
which is similar to that observed with Hemopure. In studies of human and
murine bone marrow, Optro stimulated the proliferation of burst-forming units
erythroid (BFU-E), and was able to overcome zidovudine suppression (38). Since
erythropoietin stimulates a later stage of RBC development, the colony-forming
uniterythroid (CFU-E), their effects should potentiate one another.
Early trials of Optro in end-stage renal disease, surgical blood loss, and
ANH were conducted, and a larger-scale trial of the use of this product as an RBC
substitute for intraoperative blood loss was mounted. All clinical trials with this
product were halted when the parent company, Baxter, terminated their trials with
HemAssist.
Advantages Disadvantages
but for this application, a spherical structure delimited by a bilamellar lipid mem-
brane, as shown in Figure 2, serves as a Trojan horse for transporting exogenously
supplied hemoglobin through the circulation. Liposome-encapsulated hemoglo-
bin (LEH) offers a number of potential advantages over perfluorocarbon- and
hemoglobin-based RBC substitutes, which are listed in Table 8. Many of the
problems created by the presence of cell-free hemoglobin in the plasma can be
averted, among them the need for crosslinking or other means of protecting
hemoglobin from clearance. The native oxyhemoglobin dissociation curve could
easily be preserved by encapsulating 2,3-DPG along with hemoglobin in the
liposome, and methemoglobin reductase could be added to limit autoxidation.
Encapsulated hemoglobin would presumably have minimal vasoactive effects.
The primary disadvantage of this approach lies in the complexity of
the technology for making liposomes and the difficulty of producing uniform
preparations, which might also have the effect of driving up the cost of produc-
tion. In addition, the delivery of large volumes of LEH raises the question of the
safety and particularly the impact of loading the RES with exogenous phos-
pholipids. In animal systems, several transient effects have been noted following
Hemoglobin
Phospholipids
Figure 2 Liposome-encapsulated hemoglobin (LEH).
Red Blood Cell Substitutes 561
Advantages Disadvantages
tolerance for their side effects diminishes. In addition, cost becomes an increas-
ingly important factor as the safety advantage shrinks. For the products based on
human hemoglobin, another possible limiting factor may be the availability of
source material. The supply of outdated RBC originating from volunteer donors
is presently only great enough to provide hemoglobin sufficient to supplant the
RBC supply by about 5%. Other sources of human hemoglobin would have to be
sought. The development of the technology to collect multiple units of RBCs at
one time may enhance collections, but probably not enough to significantly
augment the supply. One possible solution would be to permit collection of red
cells for further manufacture from donors who might not otherwise meet the usual
criteria or who might be paid. Blood donor centers, finding the demand for their
RBC units decreasing, may begin to develop a second category of source
hemoglobin donors, as well as emphasizing apheresis collections of platelets
and plasma. Obtaining source material may not present the same difficulties
for substitutes based on perfluorocarbons, animal hemoglobin, or recombinant
hemoglobin, assuming, for the latter, that adequate production scale-up can
be achieved.
RBC substitutes may also find a niche in the developing world where the
majority of transfusions are for acute anemia. In countries where bloodborne
pathogens are endemic, substitutes may represent a substantially safer alternative
to donor RBC. In some situations, a substitute may be less expensive to supply
than developing and maintaining the infrastructure required for a volunteer donor
blood system.
VI. CONCLUSION
In addition to the approaches taken to developing clinically useful RBC substi-
tutes described above, a number of other means of enhancing oxygen delivery,
listed in Table 9, are being explored. Most of these are much farther from clinical
studies than the products already mentioned, and one promising approach,
obtaining human hemoglobin from transgenic pigs, is not being actively pursued
by its developer.
The animal and human studies of the materials originally conceived of as
RBC substitutes have led to an appreciation for the unique and sometimes
unexpected properties of these products and has suggested applications well
beyond the role of donor red cells. Some of these potential applications are listed
in Table 10.
The search for a RBC substitute is reaching a critical juncture; the final
demonstration of safety and efficacy in broad-based phase III clinical trials.
Demonstrating the efficacy of an RBC substitute is not entirely straightforward,
since there is no uniform or generally accepted method for assessing the efficacy
of a RBC transfusion (44). Replacement of allogeneic red cell transfusions and
reduction of mortality are likely to be the criteria used to judge the effectiveness
of the RBC substitute for transfusion applications.
The demonstration of safety also faces some challenges. The risks of the
transfusion-transmitted infections, which gave such impetus to the development
of RBC substitutes, have become extremely low. The aggregate risk for the
transmission of human immunodeficiency virus (HIV), hepatitis B virus (HBV),
hepatitis C virus (HCV), and human T-lymphotropic virus type I (HTLV-I) was
estimated to be 1:34,000 units in 1996 (45) and may be even lower at present.
Fatal transfusion events are even more rare (46). The RBC substitutes will have
to exhibit exemplary side effect profiles to provide this degree of safety. They
will, however, have the advantage of being less susceptible to the emergence of
new infectious agents in the blood supply. This fear of unknown but dreaded
outcomes, which tends to exaggerate the apparent risks of donor blood, may also
add to the perceived relative safety of a substitute.
Despite some difficulties, it is likely that one or more of these oxygen-
carrying materials will be licensed for clinical use within the next few years,
either as an RBC substitute or for some other application. At that point, it will
have taken a century from the time that the discovery of the ABO blood groups
made red cell transfusion feasible to the time when a RBC substitute becomes a
clinical reality.
REFERENCES
1. Intaglietta M, Johnson PC, Winslow RM. Microvascular and tissue oxygen distribu-
tion. Cardiovasc Res 1996; 32:632643.
2. Karmaker N, Dhar P. Effect of steady shear stress on fluid filtration through the rabbit
arterial wall in the presence of macromolecules. Clin Exp Pharmacol Physiol 1996;
23:299304.
3. Gould SA, Rosen AL, Sehgal LR, Sehgal HL, Langdale LA, Krause LM, Rice CL,
Chamberlin WH, Moss GS. Fluosol-DA as a red cell substitute in acute anemia. N
Engl J Med 1986; 314:16531656.
4. Kerins DM. Role of perfluorocarbon Fluosol-DA in coronary angioplasty. Am J Med
Sci 1994; 307:218221.
5. Andre M, Nelson T, Mattrey R. Physical and acoustical properties of perfluorooctyl-
bromide, an ultrasound contrast agent. Invest Radiol 1990 25:983987.
6. Braun RD, Linsenmeier RA, Goldstick TK. New perfluorocarbon emulsion improves
tissue oxygenation in cat retina. J Appl Physiol 1992; 72:19601968.
7. Hogan MC, Kurdak SS, Richardson RS, Wagner PD. Partial substitution of red blood
cells with free hemoglobin solution does not improve maximal O2 uptake of working
in situ dog muscle. Adv Exp Med Biol 1994; 361:375378.
8. Keipert PE, Faithfull NS, Roth DJ, Bradley JD, Batra S, Jochelson P, Flaim KE.
Supporting tissue oxygenation during acute surgical bleeding using a perfluorochemi-
cal-based oxygen carrier. Adv Exp Med Biol 1996; 388:603609.
9. Flaim SF, Hazard DR, Hogan J, Peters RM. Characterization and mechanism of
side-effects of Imagent BP (highly concentrated fluorocarbon emulsion) in swine.
Invest Radiol 1991; 26(suppl):S122S128.
10. Suit H. Tumor oxygenation and radiosensitivity. In: Winslow RM, Vandegriff KD,
Intaglietta M, eds. Blood Substitutes: Physiological Basis of Efficacy. Boston:
Birkhuser, 1995:187199.
11. Rockwell S, Kelley M, Irvin CG, Hughes CS, Yabuki H, Porter E, Fischer JJ.
Preclinical evaluation of Oxygent (TM) as an adjunct to radiotherapy. Biomater Artif
Cells Immobil Biotechnol 1992; 20:883893.
12. Leach CL, Greenspan JS, Rubenstein DS, Shaffer TH, Wolfson MR, Jackson JC,
DeLemos R, Fuhrman BP. Partial liquid ventilation with perflubron in premature
infants with severe respiratory distress syndrome. N Engl J Med 1996; 335:761767.
13. Blauth CI, Arnold JV, Schulenberg WE, McCartney AC, Taylor KM. Cerebral
Red Blood Cell Substitutes 565
of diaspirin crosslinked and stroma reduced hemoglobin. Crit Care Med 1996;
24:137147.
30. Schultz SC, Powell CC, Burris DG, Nguyen H, Jaffin J, Malcolm DS. The efficacy
of diaspirin crosslinked hemoglobin solution resuscitation in a model of uncontrolled
haemorrhage. J Trauma 1994; 37:408412.
30a. Sloan EP, Koenigsberg M, Gens D, Cipolle M, Runge J, Mallory MNRG Jr. Diaspirin
cross-linked hemoglobin (DCLHb) in the treatment of severe traumatic hemorrhagic
shock. A randomized controlled efficacy trial. JAMA 1999; 282:18571864.
31. Conover CD, Malatesta P, Lejeune L, Chang CL, Shorr RG. The effects of hemodilu-
tion with polyethylene glycol bovine hemoglobin (PEG-Hb) in a conscious porcine
model. J Investig Med 1996; 44:238246.
32. Nho K, Linberg R, Johnson M, Gilbert C, Shorr R. PEG-hemoglobin: an efficient
oxygen delivery system in the rat exchange transfusion and hypovolemic shock
models. Art Cells, Blood Subs Immob Biotech 1996; 22:795803.
33. Hughes GS, Francom SF, Antal EJ, Adams WJ, Locker PK, Yancey EP, Jacobs EE
Jr. Effects of a novel hemoglobin-based oxygen carrier on percent oxygen saturation
as determined with arterial blood gas analysis and pulse oximetry. Ann Emerg Med
1996; 27:164169.
34. Hughes GS, Yancey EP, Albrecht R, Locker PK, Francom SF, Orringer EP, Antal EJ,
Jacobs EE Jr. Hemoglobin-based oxygen carrier preserves submaximal exercise
capacity in humans. Clin Pharmacol Ther 1995; 58:434443.
35. Hughes GS, Francom SF, Antal EJ, Adams WJ, Locker PK, Yancey EP, Jacobs EE
Jr. Hematologic effects of a novel hemoglobin-based oxygen carrier in normal male
and female subjects. J Lab Clin Med 1995; 126:444451.
36. Looker D, Abbott-Brown D, Cozart P, Durfee S, Hoffman S, Mathews A, Miller-
Roehrich J, Shoemaker S, Trimble S, Fermi G, Komiyama N, Nagai K, Stetler GL.
A human recombinant hemoglobin designed for use as a blood substitute. Nature
1992; 356:258260.
37. Murray JA, Ledlow A, Launspach J, Evans D, Loveday M, Conklin JL. The effects
of recombinant human hemoglobin on esophageal motor functions in humans.
Gastroenterology 1995; 109:12411248.
38. Schick MR, Rosenthal GJ. A comparative evaluation of hemin and recombinant
hemoglobin in the stimulation of erythropoietic precursors (abstr). Blood 1996;
88(suppl 1):104b.
39. Rudolph AS. Encapsulation of hemoglobin in liposomes. In: Winslow RM,
Vandegriff KD, Intaglietta M, eds. Blood Substitutes: Physiological Basis of Efficacy.
Boston: Birkhuser, 1995:90104.
40. Szebeni J, Wassef NM, Rudolph AS, Alving CR. Complement activation in human
serum by liposome-encapsulated hemoglobin: the role of natural anti-phospholipid
antibodies. Biochem Biophys Acta 1996; 1285:127130.
41. Burkard ME, Van Liew HD. Oxygen transport to tissue by persistent bubbles: theory
and simulations. J Appl Physiol 1994; 77(6):28742878.
42. Sakai H, Hamada K, Takeoka S, Nishide H, Tsuchida E. Physical properties of
hemoglobin vesicles as red cell substitutes. Biotechnol Prog 1996; 12:119125.
Red Blood Cell Substitutes 567
43. ODonnell JK, Martin MJ, Logan JS, Kumar R. Production of human hemoglobin in
transgenic swine: an approach to a blood substitute. Cancer Detect Prev 1993;
17:307312.
44. Fratantoni J. Demonstration of efficacy of a therapeutic agent. In: Winslow RM,
Vandegriff KD, Intaglietta M, eds. Blood Substitutes: Physiological Basis of Efficacy.
Boston: Birkhuser, 1995:2024.
45. Schreiber GB, Busch MP, Kleinman SH, Korelitz JJ, for the Retrovirus Epidemiology
Donor Study. The risk of transfusion-transmitted viral infections. NEJM 1996;
334:16851690.
46. Linden JV, Tourdalt MA, Scribner CL. Decrease in frequency of transfusion fatalities
(letter). Transfusion 1997; 37:243244.
25
Professional Standards and
Voluntary Accreditation
Jay E. Menitove
Community Blood Center of Greater Kansas City, Kansas City, Missouri
Hillary V. Schaeffler
American Association of Blood Banks, Bethesda, Maryland
569
570 Menitove and Schaeffler
all Standards quality concepts are consistent, and the Communications Subcom-
mittee disseminates guidance related to the standards. The program units are
responsible for creating or revising the technical/scientific standards, which are
specific for each standard-setting activity (Fig. 1).
In 1997, an Accreditation Program Committee was created to oversee all
accreditation activities. This committee comprises a chairperson, chairs of two
subcommittees (Quality Systems Subcommittee and Education Advisory Sub-
committee), and chairs of seven specialty program units (Donor Centers, Trans-
fusion Services, Hematopoietic Progenitor Cell Services, Immunohematology
Reference Laboratories, Parentage Testing Laboratories, Perioperative Collection
and Transfusion, and SBB Schools). The Quality Systems Subcommittee works
with the Accreditation Program Committee to fully implement quality principles
into AABBs Accreditation Program and to provide quality assessment tools for
AABBs institutional members. The Education Advisory Subcommittee serves as
the educational advisory group and coordinator for the Accreditation Programs
educational and training activities. Each program unit oversees and coordinates
the specific program unit activities, including coordinating and reviewing the
program assessment tools and assessor guidance (Fig. 2).
I. STANDARDS PROGRAM
The AABB entered the voluntary standard-setting arena in 1958 when it pub-
lished the first edition of Standards for a Blood Transfusion Service, the purpose
of which was to improve the quality and safety of human blood transfusions.
In 1990, the first edition of the Standards for Parentage Testing Laboratories was
published. In 1991, the first hematopoietic progenitor cell standards were added
to the Standards for Blood Banks and Transfusion Services, and 5 years later the
first edition of the Standards for Hematopoietic Progenitor Cells was published.
In 1999 the first edition of the Standards for Immunohematology Reference
Laboratories was published, and currently the first edition of Standards for
Perioperative Collection and Transfusion is being developed (Table 1).
Voluntary standards provide a benchmark for judging performance in a
specific field. For many years professionals in the legal, accounting, and engi-
neering fields have set minimum voluntary standards to determine a baseline for
compliance. Although higher standards of practice may be targeted, defining the
minimum provides a basis for measurement. When voluntary standards are
implemented, the public and current and potential customers may judge an
organizations compliance. Further, as international trade and communication
increase, voluntary standards form the basis for an objective method of distin-
guishing between those who implement the standards and those who do not.
The first edition of the blood bank Standards focused on technical specifi-
cations and followed a path of workflow approach for the blood bank and
Professional Standards and Voluntary Accreditation 571
Effective Revision
Standards Edition date cycle
Standards for Blood Banks and Transfusion Services 19th June 1999 18 months
Standards for Hematopoietic Progenitor Cell 2nd March 2000 18 months
Services
Standards for Immunohematology Reference 1st July 1999 2 years
Laboratories
Standards for Parentage Testing Laboratories 4th July 1999 2 years
Standards for Perioperative Collection and 1st July 2001 2 years
Transfusion
transfusion service, i.e., beginning with donor selection and ending with blood
administration. Over the last 40 years, 19 editions of blood bank/transfusion
service Standards have been published, each edition building on the technical
specifications from the prior edition.
Over the last decade, the AABB leadership and Standards committees have
recognized the importance of quality systems to ensure consistency in the
provision of appropriate components and services. In 1991, the requirement for a
program of quality assurance was incorporated into the 14th edition of Stand-
ards for Blood Banks and Transfusion Services. In 1996, a written quality plan
was required. The next year, in response to questions about what items should be
included in a quality plan, an association policy (Association Bulletin #97-4)
identified 10 Quality System Essentials (QSEs) as minimum requirements of a
quality system. The premise of the QSEs is that a quality management system
ensures that operations are controlled to produce consistent products and services.
To achieve and maintain this control, it is required that policies, processes, and
procedures are documented and implemented appropriately, and, when appropri-
ate, records of the activities must be created and maintained. Internal assessments
are conducted at regular, scheduled intervals, and if problems are identified,
corrective action plans must be developed.
In 1999, the 10 QSEs were fully incorporated into the 19th edition of
Standards (Table 2). In a further evolution, the framework of the 20th edition
of the Standards for Blood Banks and Transfusion Services, published in the fall
of 2000, is the 10 QSEs. The Standards publication is now organized into 10
sections, each section corresponding with a QSE. The first chapter is Manage-
ment, the second Resources, the third Equipment, etc. The technical requirements
appear in the appropriate chapter, creating a matrix of technical and quality
requirements. For example, instead of repeating that equipment must be main-
tained throughout the standards, the requirement is stated once in Section 3,
574 Menitove and Schaeffler
Equipment. Then the specific requirements for equipment (e.g., blood warmers,
alarms) are placed in that section. With this model, the technical standards are as
important as the quality requirements, but this system incorporates them into a
quality management system.
confidence in the standards setting process, the AABB Standards Committee base
their decisions on scientific evidence, when available, good medical practice,
technological changes, public policy issues, and government regulations and
guidelines. When there are differences of opinion, committee decisions are made
by a majority vote of the members present.
Once the revision phase of the process is completed, the Standards are
presented to the Standards Program Committee and the Board of Directors for
approval to publish as proposed Standards. Once approved, the proposed changes
are published for comment on the public section of the AABB web site for 60
days. Notice of the proposed Standards is printed in a nationally circulated
newspaper and AABB newsletters. Following the comment period, the program
unit reviews each comment, and final wording is determined. The final draft
undergoes a thorough legal, regulatory, and technical review and is submitted to
the Standards Program Committee and Board for final approval. Once the final
Standards are issued, institutional members have 60 days to implement them.
If it is determined that a new standard must be set or a current standard must
be revised before the next edition is published, a program unit may add or revise
a standard if there is scientific evidence for doing so. In this instance, a proposed
interim standard is posted for an abbreviated comment period. After consideration
of the comments and with Standards Program Committee and Board approval, an
interim standard is set and must be implemented by accredited members. In rare
circumstances, when an accelerated review and approval process is required, an
emergent standard is set with Board approval but without a comment period.
If an institution has data to prove that it can meet the intent of a standard
through another mechanism, it may apply to the relevant program unit for a
variance or an exemption. In this case, the program unit considers the request,
and if the variance is approved the Communications Subcommittee communi-
cates the decision. If an institution disagrees with a variance decision, it may
appeal the decision to a Standards Review Committee. Variances apply for one
edition of Standards, although institutions may reapply for variances.
V. ACCREDITATION PROGRAM
Following publication of the first edition of standards in 1958, AABBs Inspec-
tion and Accreditation program was developed to identify whether inspected
organizations met the requirements of the standards (a detection-oriented model).
Inspectors used a checklist during on-site inspections to determine whether the
specific requirements in the standards had been met. Following a review of the
inspectors completed checklist, summary reports of the inspections were written
by area chairs.
In 1997 a new process was developed to incorporate a systems approach
(a prevention-oriented model). Checklists transitioned to assessment tools, and
the terminology of inspectors transitioned to assessors. With this new model,
summary reports are left on-site at the conclusion of the assessment and assessors
identify nonconformances that may be an isolated incident or a systems-related
problem. Assessors evaluate both the quality and operational activities performed
within an institution. The quality system evaluation is based on the same criteria
for every facility; the operational systems are identified by the activities per-
formed by the institution. The result is that each assessment is customized for
the facility.
At the core of the accreditation program are hundreds of volunteer asses-
sors. Assessors must have appropriate expertise, participate in assessor continuing
education, and perform a minimum number of assessments each year. Assessors
and their organizations benefit from free training in quality concepts and auditing
techniques, networking with other individuals in their profession, and learning
about best practices used in other facilities. In 1999 the AABB also hired three
Professional Standards and Voluntary Accreditation 577
lead assessors who have expertise in the field and are well trained in quality and
quality assessments.
As the Food and Drug Administration, Health Care Financing Administra-
tion, and Joint Commission on Accreditation of Healthcare Organizations focus
on the importance of quality assurance and systems evaluation, the AABB is
confident that its standards and accreditation program is preparing AABB-
accredited institutions for the changing environment. Institutions that are accred-
ited by the AABB send a signal to their customers and the public that they
implement voluntary industry standards, which require control of operations
through a quality management system and implementation of the blood banking
communitys agreed-upon technical standards. After more than 40 years in the
standard-setting arena, the AABBs standards and accreditation program has
played a significant role in improving patient safety, and institutions with AABB
accreditation are respected around the world.
26
The Role of Federal Regulation in
Blood Safety
Jay S. Epstein and Mary Gustafson
U.S. Food and Drug Administration, Rockville, Maryland
I. STATUTORY HISTORY
The modern blood industry dates back more than 50 years. Establishments known
as blood banks began to appear in the 1930s, and widespread use of blood and
its derivatives began during World War II. Federal regulatory control was exerted
over blood and blood products almost from their inception based on the preexist-
ing Biologics Control Act of 1902 (also known as the Virus-Toxin Act). This law
requiring licensing of biologics was consolidated with other public health laws in
1944 to become the Public Health Service (PHS) Act. The first blood product,
Normal Human Plasma, was licensed for medical use in 1940. The first blood
bank was licensed for the manufacture of whole blood in 1946. The first
blood grouping reagent for serological testing of red blood cells was licensed in
1949. The PHS Act was implemented by the National Institutes of Health (NIH)
during the time the first blood licenses were issued. Blood was also considered
to be a drug subject to the Federal Food, Drug and Cosmetic (FDC) Act; however,
the full scope of regulatory controls available under the FDC Act was not
implemented until regulatory control for biologics was transferred from the NIH
to the U.S. Food and Drug Administration (FDA) in 1972 (1). Today, blood, blood
components, blood-derived and analogous products, and in vitro diagnostic blood
screening tests and other medical devices used in the manufacture of blood and
blood components are regulated under federal statutes implemented by FDA.
The primary statutes covering blood regulation are the PHS Act (42 USC
202 et. seq.) and the FDC Act (21 USC 302 et. seq.). Both laws have predecessors
that date to the early 1900s. Both were enacted following tragic events that
579
580 Epstein and Gustafson
motivated the U.S. Congress to insist that medicines and other medical use
products be controlled. The impetus for the enactment of the first biologics statute
was the death of several children in the fall of 1901. Those children had been
given a diphtheria antitoxin contaminated with tetanus toxin. The source of the
antitoxin was a horse that later developed tetanus. The event was deemed
preventable had controls been in place throughout the procurement and process-
ing of the antitoxin. Congress therefore enacted the Biologics Control Act, which
required the licensing of manufacturing establishments as well as the biological
products they manufactured. Thus, the Act, which later was incorporated into
Section 351 of the PHS Act, serves as the legal basis of licensing biologics (2).
The original PHS Act did not specifically address blood and blood derivative
products, but these products were regulated as being analogous to a therapeutic
serum, a category specifically included in the language of the statute. This
interpretation was challenged in 1968 in Blank v. United States (3). Mrs. Maxine
Blank had been convicted of misdemeanor charges under the PHS Act for
violations incurred in operating a commercial blood bank located in Dallas,
Texas. She was found guilty but appealed her conviction. The grounds for appeal
were based upon the absence of any reference to blood or blood products in the
statute. The court agreed with Mrs. Blank and reversed the conviction. However,
it was not the intention of Congress to omit the regulation of blood and
blood-derived products from the authority of the PHS Act, and in 1970 Con-
gress amended the Act specifically to include, blood, blood components and
derivatives (4).
The FDC Act was preceded by the Pure Food and Drug Act of 1906. This
early law was passed following disclosure of unsanitary and uncontrolled prac-
tices in the meat-packing industry in the popular book The Asphalt Jungle by
Upton Sinclair. The early law addressed the purity of foods and drugs and was
silent with respect to their safety and efficacy. The drug act was strengthened in
1938, however, after more than 100 people died after ingesting elixir of sulfanil-
amide in which the drug was dissolved in diethylene glycol, an extremely toxic
substance, rather than alcohol or water. The recodified 1938 law was the forerun-
ner of the modern FDC Act and included the requirement for safety as well as
purity (5). The law was further amended in 1962, after the thalidomide tragedy,
to include efficacy as well as safety, and again in 1976, to encompass the
regulation of medical devices. Until the Food and Drug Administration Modern-
ization Act of 1997, the only other major change to the FDC Act was strengthen-
ing of device regulation and clarification of the regulation of combination drug
and device products by the Safe Medical Device Act of 1990 (6,7).
Key provisions of the PHS Act and the FDC Act are outlined in Table 1.
Besides differing in structure, the two statutes differ in objective, scope, focus,
and enforcement mechanism. The PHS Act was enacted to promote public health
and disease prevention by assuring safe and effective vaccines and other biolog-
Federal Regulation and Blood Safety 581
Section Provision
PHS Act
351(a) Requires licenses for biological products
351(b) Prohibits false labeling
351(c) Authorizes inspections
351(a) Authorizes suspension/revocation of licenses
361 Provides authority to control spread of communicable disease
FDC Act
301, et seq Outlines prohibited acts and penalties
501 Prohibits adulteration
502 Prohibits misbranding
510 Requires registration of producers of drugs and devices
704 Authorizes inspections
ical products intended to treat and prevent diseases of humans. The FDC Act was
intended to guard the consumer against adulterated and misbranded foods, drugs,
devices, and cosmetics. The PHS Act provides for administrative actions of
license suspension and revocation as the primary remedies for violating the
statute. In contrast, the FDC Act is far more enforcement-oriented in that it
outlines prohibited acts and describes remedies that are judicial, e.g., product
seizure, prosecution, and injunction. However, since biological products under the
PHS Act concurrently are drugs or devices under the FDC Act, the enforcement
provisions of the FDC Act can be applied.
Cite Addresses
(IDE) regulations in 21 CFR, Part 812. Just as the preapproval processes differ
depending on whether the product is a drug, device, or biological drug or device,
the approval processes differ as well. Products that are deemed to be biologics
are regulated under the licensing provisions of Section 351 of the PHS Act.
Products regulated in this manner include blood and blood components, blood-
derived and related products, and in vitro tests required or recommended for
testing the blood supply. Other medical devices used to process, test, store, or
administer blood are regulated under the medical device amendments to the FDC
Act. Depending upon the classification of the device, permission to market it is
granted by approval of a premarket approval application (PMA) or clearance of
a premarket notification (510k). Blood anticoagulants, additive and rejuvenation
solutions, and colloidal plasma volume expanders are subject to the drug-approval
mechanisms under Section 505 of the FDC Act. In accordance with intercenter
agreements between the Center for Biologics Evaluation and Research (CBER),
the Center for Drug Evaluation and Research, and the Center for Devices and
Radiological Health, CBER has jurisdiction for the regulation of blood as well as
drugs and devices used in the processing of blood regardless of the regulatory
mechanism (10,11).
The majority of blood and blood-related products are licensed as biologics
under the PHS Act. Therefore, the remainder of this chapters discussion will
focus on licensing and the related regulatory requirements. Prior to the im-
plementation of the Food and Drug Administration Modernization Act of 1997,
licensing of blood products required that the product and the establishment
responsible for the manufacture of the product each be licensed. That is, separate
licenses were issued for the biological product (product license) and the facility
manufacturing the product (establishment license). Since February 1998, a single
biologics license has been issued. On July 30, 1998, FDA published a proposed
rule to amend the biologics regulations to eliminate the filing of a separate
establishment license application and a product license application in order to
market a biological product in interstate commerce. The separate license applica-
tions are being replaced by a single biologics license application (12). The impact
of this change is discussed later in this chapter.
violative of regulations are listed on the form. Further compliance action, such as
issuance of a warning letter, follows only after the results of the inspection,
including the Form 483 observations, are reviewed by compliance officers at the
district level and sometimes at the Center level. Administrative and legal actions
such as license suspension or revocation, injunction, seizure, and prosecution are
infrequent occurrences. Voluntary compliance is the usual outcome and a goal
of FDA.
Except in situations of imminent threat to health, if an establishment is
found violating any of the laws that FDA enforces, it usually is given a chance to
correct the problem voluntarily before FDA pursues an enforcement action. When
an establishment cannot or will not correct a violative situation, FDA can invoke
administrative or legal sanctions. If a firm is licensed and deficiencies present are
grounds for revocation (21 CFR 601.5), the firm can be advised that the agency
will proceed to revoke the firms license. Further, if a danger to health exists, the
firms license can be summarily suspended. Legal actions against a firm include
injunction and prosecution. Mandatory injunction (specifying corrective action),
as opposed to prohibitory injunction or license revocation (causing cessation of
operations), is the remedy most frequently sought when there is a need to maintain
the blood supplied by a firm operating out of compliance. Violative products can
be removed from the market by voluntary removal or correction of labeling by
the manufacturer. These actions are classified as recalls when FDA otherwise
would take action against the violative product, i.e., by seizure (17). See appendix
C for a summary of enforcement actions in recent years.
B. Changes in Licensing
Section 351 of the PHS Act requires that a biological license be in effect for
commercial interstate distribution of a biological product. Regulations (21 CFR,
Section 601) implementing the PHS Act in the past required that licensing be
accomplished by the application for and issuance of separate licenses for the
establishment preparing the biological product and the product itself. The require-
ment for separate licensing of an establishment and each product manufactured
has been changed. The agency has eliminated the establishment license filing for
all biological products, including blood, blood components, and derivatives (12).
The current establishment license application and multiple product license appli-
cations have been replaced with a single form. Applications for product approval
(i.e., new drug and biologics licenses) are accomplished by the submission of one
harmonized application form for all drugs and biological products, and one
biologics license approval will issue covering the biological product and its
manufacturing facilities and processes. The harmonized application (FDA Form
Federal Regulation and Blood Safety 589
D. Gene-Based Testing
In September 1994, FDA sponsored a workshop entitled Conference on the
Feasibility of Genetic Technology to Close the HIV Window in Donor Screen-
ing. The purpose of the workshop was to gather data on ways to close the
Federal Regulation and Blood Safety 591
NAT testing for the currently approved HIV-1 p24 antigen test if pool testing is
shown to be sufficiently sensitive.
April 23, 1992, the recommendation was revised to extend testing to both
products for transfusion and for further manufacturing into injectable products.
This decision coincided with FDA approval of the first multiantigen test for
antibodies to HCV, a test with sensitivity greater than 90%. A supplemental, more
specific test was licensed (available previously only as an investigational test) in
mid-1993.
Tracing of recipients who received blood components from donors later
found reactive for antibodies to HCV has been termed targeted lookback.
Although targeted lookback for HCV had been discussed in public meetings since
October 1989, it was not recommended for a variety of reasons. Most importantly,
in the absence of supplemental testing, the positive predictive value of the first
HCV screening test was low and the significance of a reactive test in terms of
infectivity was not known. Additionally, few HCV-infected individuals were
eligible for investigational treatment of their liver disease, and the long-term
effect of treatment was unknown.
Over time, it became known that individuals reactive for anti-HCV in a
supplemental assay are likely to be chronically infected with HCV. More specif-
ically, in studies of blood donors, 7395% of supplemental testpositive and
1421% of supplemental testindeterminate blood donors had detectable HCV
RNA by PCR (2931). It also is now recognized that negative or unscreened units
from donors later found reactive for anti-HCV may have been contaminated with
HCV. In parallel with these improved understandings, there have been im-
provements in the management and treatment of HCV infections that were
recognized by publication of treatment guidelines developed at a NIH Consensus
Conference in March 1997. Driven in large measure by congressional concerns
over the emerging epidemic of liver disease from chronic HCV infections, the
issue of targeted lookback for HCV was brought before the PHS ACBSA. The
Committee discussed the topic at its April 2425, 1997, meeting and its August
1112, 1997, meeting. After careful consideration, the Committee recommended
that the PHS initiate a targeted lookback program extending back 10 years to
identify recipients of previously donated units from donors who have tested
positive for antibody to HCV following screening by a multiantigen screening
test [enzyme immunoassay (EIA)] since 1992.
In March 1998 and September 1998, FDA issued guidance consistent with
this recommendation. At public meetings on November 24, 1998, and January 28,
1999, the PHS ACBSA reconsidered the issue of recipient notification related to
repeatedly reactive results on the EIA 1.0 test. Following acceptance by the
committee, FDA issued guidance on June 22, 1999, to replace earlier guidances
(32). This current guidance provides for lookback to EIA 1.0 testing and also
recommends that the search of records of prior donations from donors with
repeatedly reactive screening tests for HCV extend back indefinitely to the extent
that electronic or other readily retrievable records exist.
Federal Regulation and Blood Safety 595
REFERENCES
1. Solomon JM. The evolution of the current blood banking regulatory climate. Trans-
fusion 1994; 34:272277.
2. Noguchi PD. From Jim to Gene and beyond: an odyssey of biologics regulation. Food
Drug Law 1996; 51:367373.
3. Blank v. United States. Federal Reporter, 2d Series 1968; 400:302306.
4. Public Law No. 91-515, 58 Stat. 702 (1970).
5. Beatrice MG. Regulation, licensing and inspection of biological products. Phar-
maceut Eng 1991; 10:2935.
596 Epstein and Gustafson
6. The Food and Drug Administration Modernization Act of 1997. Public Law No.
105-115, 1997.
7. Compilation of Laws Enforced by the U.S. Food and Drug Administration and
Related Statutes. Vol 1. Washington, DC: U.S. Government Printing Office, 1996.
8. Leveton LB, Sox HC Jr., Stoto MA, eds. HIV and the Blood Supply: An Analysis of
Crisis Decision Making. Washington, DC: National Academy Press, 1995.
9. The Food and Drug Administrations Development, Issuance, and Use of Guidance
Documents. Fed Reg 1997; 62:89618972.
10. Intercenter Agreement Between the Center for Drug Evaluation and Research and the
Center for Biologics Evaluation and Research. Rockville, MD: Food and Drug
Administration, 1991.
11. Intercenter Agreement Between the Center for Drug Evaluation and Research and the
Center for Devices and Radiological Health. Rockville, MD: Food and Drug Admin-
istration, 1991.
12. Biological Products Regulated under Section 351 of the Public Health Service
Act: Implementation of Biologics License Elimination of Establishment License
and Product License. Fed Reg 1999; 64:S6441S6454.
13. Changes to an approved application. Fed Reg 1997; 62:3989039906.
14. Biological products; reporting of errors and accidents in manufacturing: proposed
rule. Fed Reg 1997; 62:4964249648.
15. Team Biologics: A Plan for Reinventing FDAs Ability to Optimize Compliance of
Regulated Biologics Industry. Rockville, MD: Food and Drug Administration, 1997.
16. Compliance Program Guide for Plasma Fractionation Facilities. Rockville, MD: Food
and Drug Administration, 1997.
17. Tourault MA. Modern principles of blood banking compliance with Food and Drug
Administration regulations, In: Harmening DM, eds. Modern Blood Banking and
Transfusion Practices. 3rd ed. Philadelphia: F.A. Davis Company 1994:288302.
18. Blood Supply: FDA Oversight and Remaining Issues of Safety. Washington, DC:
General Accounting Office, 1997; PEMD-97-1.
19. Recommendations for Donor Screening with a Licensed Test for HIV-1 Antigen.
Food and Drug Administration, 1995.
20. Schreiber GB, Busch MP, Kleinman SH, Korelitz JJ. The risk of transfusion-
transmitted viral infections. N Engl J Med 1996; 334:16851690.
21. Precautionary Measures to Further Reduce the Possible Risk of Transmission of
Creutzfeldt-Jakob Disease by Blood and Blood Products. Rockville, MD: Food and
Drug Administration, 1995.
22. Revised Precautionary Measures to Reduce the Possible Risk of Transmission of
Creutzfeldt-Jakob Disease (CJD) by Blood and Blood Products. Fed Reg 1997;
62:4969449695.
23. Will RG, Ironside JW, Zeidler M. A new variant of Creutzfeldt-Jakob disease in the
U.K. Lancet 1996; 347:921925.
24. Sullivan MT, Schonberger LB, Kessler D, Williams A, Dodd R. Creutzfeldt-Jakob
disease (CJD) investigational lookback study. Transfusion 1997; 37(suppl):2S.
25. Heye N, Hensen S, Muller N. Creutzfeldt-Jakob disease and blood transfusion.
Lancet 1994; 343:298299.
26. Change to the Guidance Entitled Revised Precautionary Measures to Reduce the
Federal Regulation and Blood Safety 597
As a service to the public, FDAs Center for Biologics Evaluation and Research
(CBER) provides information in a variety of ways.
Internet
CBERs Internet site is located at: http://www.fda.gov/cber/
The site contains a myriad of information including:
Product information, e.g., recall/withdrawal/safety issues, product
approvals, information sheets, adverse event reporting information
(www.fda.gov/cber/products.htm);
On line documents, e.g., guidelines/guidances, product approval docu-
ments, establishment and product files, Federal Register notices, informa-
tion sheets, letters to industry and healthcare providers, memoranda to
blood establishments, points to consider, and general and administrative
information about CBER (www.fda.gov/cber/publications.htm);
Documents available electronically under the Freedom of Information Act
(FOI) and how to submit an FOI request (www.fda.gov/cber/efoi.htm);
Information on FDA Modernization Act (www.fda.gov/cber/fdama.htm)
and Prescription Drug User Fee Act (www.fda.gov/cber/pdufa.htm)
Callers outside the U.S. must call this system from a FAX machine with a
touch-tone telephone attached or built in.
When prompted to enter your FAX number, please enter the entire 10 digit
number, including the 3 digit area code, even if you are local to Rockville, MD.
Do not include a 1 for long distance dialing.
To obtain a complete list of documents available from this system,
select document 9999
To obtain a list of documents added to the system in the last 30 days,
select document 9998
To obtain a list of Recall/Withdrawal/Safety notifications, select docu-
ment 9997
Up to 5 documents can be ordered at a time. Please enter the document
number(s) listed on the index.
E-Mail
Consumer questions about biological products can be sent by e-mail
to: OCTMA@CBER.FDA.GOV
Manufacturers assistance questions can be sent by e-mail to: MATT
@CBER.FDA.GOV
Documents can be requested by e-mail from: CBER_INFO@CBER.
FDA.GOV
Telephone
Voice Information System
Direct access to Consumer Safety Officers or Public Affairs Specialists:
1-800-835-4709 or 301-827-1800
Blood and Plasma Products Information
Recall and market withdrawal notices for fractionated plasma products:
1-888-CBER-BPI or 301-827-4604
Division of Blood Applications 301-827-3543 (for questions about
your application)
Blood and Plasma Branch (fax: 301-827-2857)
Regulatory Project Management Branch (fax: 301-827-3534)
Devices Review Branch (fax: 301-827-3535)
FPRECALL
Members of this list will receive notices of recalls and market
withdrawals of fractionated products.
BLOODINFO
Members of this list will receive all FPRECALL notices and other
blood-related documents.
CBERINFO
Members of this list will receive FPRECALL and BLOODINFO
documents, and notification of ALL new documents, including
guidelines, points to consider, Whats New on CBERs web site, and
other CBER information.
Printed Copy
Single copies of documents are available. Requests may be sent in writing to:
Documents may also be obtained by calling CBERs Voice Information System at:
1-800-835-4709 or 301-827-1800
For a complete list of guidelines, guidance, points to consider and other docu-
ments available in hard copy, request document D9001 (hard copy ID number)
or document 9001 from the FAX Information System.
For a complete list of memoranda and related documents pertaining to blood and
blood products available in hard copy, request document D9002 (hard copy ID
number ) or document 9002 from the FAX Information System.
Federal Regulation and Blood Safety 601
APPENDIX B
death __________________ congenital anomaly 4. Diagnosis for use (indication) 5. Event abated after use
stopped or dose reduced
(mo/day/yr)
required intervention to prevent #1
life-threatening permanent impairment/damage #1 yes no doesn't
apply
hospitalization initial or prolonged other: #2
___________________
6. Lot # (if known) 7. Exp. date (if known)
#2 yes no doesn't
apply
3. Date of 4. Date of
#1 #1 8. Event reappeared after
event this report
(mo/day/yr) (mo/day/yr) reintroduction
#2
5. Describe event or problem #2
#1 yes no doesn't
apply
9. NDC # for product problems only (if known)
#2 yes no doesn't
apply
10. Concomitant medical products and therapy dates (exclude treatment of event)
2. Type of device
________________
5. Expiration date
6. (mo/day/yr)
model #________________________________________
7. If implanted, give date
6. Relevant tests/laboratory data, including dates catalog #_______________________________________ (mo/day/yr)
serial # ________________________________________
8. If explanted, give date
lot # ___________________________________________ (mo/day/yr)
other #
9. Device available for evaluation? (Do not send to FDA)
yes no returned to manufacturer on _________________
(mo/day/yr)
10. Concomitant medical products and therapy dates (exclude treatment of event)
E. Initial reporter
1. Name & address phone #
Medication and Device Submission of a report does not constitute U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES
Public Health Service Food and Drug Administration
an admission that medical personnel, user
Experience Report facility, distributor, manufacturer or product
(continued) caused or contributed to the event.
Refer to guidelines for specific instructions Page ____ of ____ FDA Use Only
no
(mo/day/yr)
home diagnostic facility
nursing home ambulatory recall notification
initial use of device
13. Report sent to manufacturer?
outpatient
surgical facility reuse
repair inspection
yes ___________________
treatment facility unknown
no (mo/day/yr) other: replace patient monitoring
9. If action reported to FDA under
specify
14. Manufacturer name/address relabeling modification/ 21 USC 360i(f), list correction/removal
adjustment reporting number:
other:
G. All manufacturers
1. Contact office name/address (& mfring site for devices) 2. Phone number
3. Report source
(check all that apply)
foreign
study
literature
consumer
health
4. Date received by manufacturer 5. professional
(mo/day/yr) (A) NDA # ___________
user facility
6. If IND, protocol #
IND # ___________ company
representative
PLA # ___________
distributor
pre-1938 yes
other:
7. Type of report
(check all that apply) OTC
product
yes
5-day 15-day
8. Adverse event term(s)
10-day periodic
The public reporting burden for this collection of information has been estimated to average one- DHHS Reports Clearance Office An agency may not conduct or sponsor,
hour per response, including the time for reviewing instructions, searching existing data sources, Paperwork Reduction Project (0910-0291) and a person is not required to respond to,
Please DO NOT RETURN this
gathering and maintaining the data needed, and completing and reviewing the collection of infor- Hubert H. Humphrey Building, Room 531-H a collection of information unless it displays form to this address.
mation. Send comments regarding this burden estimate or any other aspect of this collection of 200 Independence Avenue, S.W. a currently valid OMB control number.
information, including suggestions for reducing this burden to: Washington, D.C. 20201
APPENDIX C
Revocationsa 5 0 3 5 5
Suspensions 3 1 3 3 0
Injunctions 0 1 3 1 0
Seizures 0 0 3 0 1
Warning letters 19 21 46 31 27
aIncludes Notices of Intent to Revoke.
Recalls Classified
Type of
product FY95 FY96 FY97 FY98 FY99
APPENDIX D
APPLICANT INFORMATION
NAME OF APPLICANT DATE OF SUBMISSION
TELEPHONE NO. (Include Area Code) FACSIMILE (FAX) Number (Include Area Code)
APPLICANT ADDRESS (Number, Street, City, State, Country, ZIP Code or Mail Code, AUTHORIZED U.S. AGENT NAME & ADDRESS (Number, Street, City, State,
and U.S. License number if previously issued): ZIP Code, telephone & FAX number) IF APPLICABLE
PRODUCT DESCRIPTION
NEW DRUG OR ANTIBIOTIC APPLICATION NUMBER, OR BIOLOGICS LICENSE APPLICATION NUMBER (If previously issued)
ESTABLISHED NAME (e.g., Proper name, USP/USAN name) PROPRIETARY NAME (trade name) IF ANY
APPLICATION INFORMATION
APPLICATION TYPE
(check one) NEW DRUG APPLICATION (21 CFR 314.50) ABBREVIATED NEW DRUG APPLICATION (ANDA, 21 CFR 314.94)
BIOLOGICS LICENSE APPLICATION (21 CFR Part 601)
TYPE OF SUBMISSION (check one) ORIGINAL APPLICATION AMENDMENT TO A PENDING APPLICATION RESUBMISSION
IF A SUPPLEMENT, IDENTIFY THE APPROPRIATE CATEGORY CBE CBE-30 Prior Approval (PA)
REASON FOR SUBMISSION
PROPOSED MARKETING STATUS (check one) PRESCRIPTION PRODUCT (Rx) OVER THE COUNTER PRODUCT (OTC)
NUMBER OF VOLUMES SUBMITTED THIS APPLICATION IS PAPER PAPER AND ELECTRONIC ELECTRONIC
ESTABLISHMENT INFORMATION (Full establishment information should be provided in the body of the Application.)
Provide locations of all manufacturing, packaging and control sites for drug substance and drug product (continuation sheets may be used if necessary). Include name,
address, contact, telephone number, registration number (CFN), DMF number, and manufacturing steps and/or type of testing (e.g. Final dosage form, Stability testing)
conducted at the site. Please indicate whether the site is ready for inspection or, if not, when it will be ready.
Cross References (list related License Applications, INDs, NDAs, PMAs, 510(k)s, IDEs, BMFs, and DMFs referenced in the current application)
PAGE 1
Federal Regulation and Blood Safety 605
This application contains the following items: (Check all that apply)
1. Index
2. Labeling (check one) Draft Labeling Final Printed Labeling
B. Samples (21 CFR 314.50 (e)(1); 21 CFR 601.2 (a)) (Submit only upon FDAs request)
C. Methods validation package (e.g., 21 CFR 314.50(e)(2)(i); 21 CFR 601.2)
5. Nonclinical pharmacology and toxicology section (e.g., 21 CFR 314.50(d)(2); 21 CFR 601.2)
6. Human pharmacokinetics and bioavailability section (e.g., 21 CFR 314.50(d)(3); 21 CFR 601.2)
12. Case report forms (e.g., 21 CFR 314.50 (f)(2); 21 CFR 601.2)
13. Patent information on any patent which claims the drug (21 U.S.C. 355(b) or (c))
14. A patent certification with respect to any patent which claims the drug (21 U.S.C. 355 (b)(2) or (j)(2)(A))
( )
Public reporting burden for this collection of information is estimated to average 24 hours per response, including the time for reviewing
instructions, searching existing data sources, gathering and maintaining the data needed, and completing and reviewing the collection of
information. Send comments regarding this burden estimate or any other aspect of this collection of information, including suggestions for reducing
this burden to:
Department of Health and Human Services Food and Drug Administration
Food and Drug Administration CDER, HFD-94 An agency may not conduct or sponsor, and a
CBER, HFM-99 12420 Parklawn Dr., Room 3046 person is not required to respond to, a collection
1401 Rockville Pike Rockville, MD 20852 of information unless it displays a currently valid
Rockville, MD 20852-1448 OMB control number.
FORM FDA 356h (4/00)
PAGE 2
Index
Page numbers in italic indicate figures or tables.
607
608 Index