J Antimicrob Chemother 2016; 71: 2071 – 2074
doi:10.1093/jac/dkw083 Advance Access publication 10 April 2016
Bacteriophage therapy: a regulatory perspective
Eric Pelfrene1*, Elsa Willebrand1, Ana Cavaleiro Sanches2, Zigmars Sebris3 and Marco Cavaleri1
1
*Corresponding author. Tel: +44-20-3660-8593; Fax: +44-20-3660-5515; E-mail: eric.pelfrene@ema.europa.eu
Despite the recognized problem of antibiotic multidrug resistance, very few antibacterial agents with new
mechanisms of action are under development. Bacteriophage therapy could offer one alternative strategy to
mitigate this challenge. Although widely used throughout the 20th century in Eastern Europe and the former
Soviet Union, this potential therapy has not yet been investigated according to rigorous scientific standards.
This paper reports on a multistakeholder meeting held at the EMA, which outlined the existing regulatory framework to which such therapy should adhere and reviewed the current obstacles and shortcomings in scientific
development for bacteriophage therapy.
Introduction
Lytic bacteriophage (phage) therapy may be a promising intervention for the treatment of bacterial infection, either in addition
to or as an alternative to conventional antibacterial treatments.
The investigation of phage therapy is particularly pertinent considering the emerging global threat of antimicrobial resistance.
Co-discovered by Twort1 and d’Hérelle,2 phages have been
used in medicine since 1919, a decade before the discovery of
antibiotics. The first scientific article describing bacteriophage
therapy was published in 1921.3 Although bacteriophage therapy
was largely replaced by antibiotics in Western countries after the
Second World War, it remained a popular treatment throughout
the 20th century in Eastern Europe (Poland) and in the former
Soviet Union (Georgia, Russia). As a result, extensive observational
data have been gathered for various infection types.4 More
recently, some data have been emerging from human trials conducted to modern standards5,6; however, there is still a lack of
data to prove their efficacy and safety.
Phage therapy has been conceived either as a ‘personalized’
treatment option administered to an individual patient (as a specific, tailored bacteriophage following isolation and identification
of the causative pathogen) or as a ‘ready to use’ cocktail that
comprises several different strains of phages targeting one or
more pathogenic bacterial species. Potential advantages and
drawbacks for the use of natural phages and genetically modified
phages have been described.7 Some investigators have claimed
that the available regulatory framework potentially hampers successful commercialization of phage therapy and access by
patients, particularly for the personalized treatment option.8,9
Aim of the EMA workshop
In June 2015, the EMA invited representatives from industry, academia, regulatory authorities and European legislators for a
workshop on the therapeutic use of bacteriophages. This workshop was part of the EMA’s commitment to explore new therapeutic options for difficult-to-treat infections, including those
due to MDR bacteria. The aim of the meeting was to facilitate
the development of bacteriophage therapy by reviewing the scientific and regulatory aspects related to this potential therapeutic
alternative. It covered general regulatory considerations and
quality aspects of phage therapy as well as the state of the art
of clinical development to date. Participants explored how regulators could ease the path towards mainstream clinical use without
compromising on standards as laid down in current legislation on
requirements for proof of quality, safety and efficacy. The recording of the workshop and various presentations have been made
available on the EMA web site event page (http://www.ema.
europa.eu/ema/index.jsp?curl=pages/news_and_events/events/
2015/05/event_detail_001155.jsp&mid=WC0b01ac058004d5c3).
Regulatory considerations
At the workshop, it was clarified that bacteriophage therapy falls
under the scope of the existing European regulatory framework on
biological medicinal products, as outlined in Directive 2001/83/
EC.10 Hence, in general, a marketing authorization is required
before these products can be used in patients, based on detailed
pharmaceutical, preclinical and clinical documentation, in accordance with the directive. However, the legislation foresees some
exceptions that would not require a marketing authorization.
Amongst others, these exceptions include new medicines that
are intended for research and development trials, named-patient
use programmes and compassionate use programmes.
Further, natural phages are not considered advanced therapy
medicinal products (ATMPs), and there are no precedents of bacteriophages expressing recombinant nucleic acid being classified
as a gene therapy product (a subcategory of ATMPs). However, if
bacteriophages expressing recombinant nucleic acid were to be
# The Author 2016. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved.
For Permissions, please e-mail: journals.permissions@oup.com
2071
Downloaded from https://academic.oup.com/jac/article-abstract/71/8/2071/2237822 by guest on 14 June 2020
Office of Anti-infectives and Vaccines, Human Medicines Evaluation Division, European Medicines Agency, London, UK; 2Quality Office,
Human Medicines Evaluation Division, European Medicines Agency, London, UK; 3Regulatory Affairs Office, Human Medicines Research and
Development Support Division, European Medicines Agency, London, UK
Review
Quality aspects
The main quality prerequisites of a pharmaceutical product and
particularly a biological product were discussed during the EMA
meeting. Manufacturers mentioned that in the absence of specific
2072
quality guidelines for phage products, they generally adhere to
existing guidance for biotechnology and biological products.
Participants discussed and agreed that the following parameters need careful consideration in the production process of
a phage product: bacteriophage identification, potency and
biological activity of the preparation, product purity, stability and
storage conditions as well as control of sterility.
Bacteria and phage bank systems, which typically consist of
master cell banks and working cell banks, need to be established.
The generation and characterization of the banks should be
performed in accordance with the general principles laid down
in the Committee for Medicinal Products for Human Use
and International Conference on Harmonisation of Technical
Requirements for Registration of Pharmaceuticals for Human
Use (CHMP/ICH) Q5D Guideline ‘Quality of biotechnological products: derivation and characterization of cell substrates used for
production of biotechnological/biological products’.15 The banked
phages and bacteria should be characterized for relevant phenotypic and genotypic markers so that amongst other parameters,
identity, viability, potency and purity are ensured.
With regard to the phages, it is recommended to use only
lytic phages (and no temperate phages) unable to transfer host
bacterial DNA (potential virulence genes) into non-targeted
bacteria [transduction or phage-mediated horizontal gene transfer
(HGT)]. Following lysogenic induction, temperate phages may
transfer fragments of DNA containing toxin-encoding or antibiotic
resistance-mediating genes, and thus temperate phages could
produce new pathogenic strains. Therefore, it is important to
ensure that therapeutic phages do not carry out generalized transduction and do not possess gene sequences having significant
homology with major antibiotic resistance genes, genes for
phage-encoded toxins or genes for other bacterial virulence factors. Nevertheless, it was pointed out that currently it is not feasible
to exclude the possibility of low levels of generalized transduction
by therapeutic phages into any of the infecting and commensal
bacteria present in the patient. Moreover, it was mentioned that
the use of phages that mediate some random general transduction
might be considered in certain circumstances (science- and riskbased decision, taking into consideration the patients’ needs).16
Concerning the host bacteria for manufacturing, it was mentioned that non-lysogenic strains containing few phages or
other phage-like elements of genetic exchange should be used.
Nevertheless, it was emphasized that most often it will not be
possible to find or quickly generate a suitable host bacterium
that is free of prophages or phage-like elements.
An example was given in which several active substances
(phages) can be produced in parallel to be combined into a drug
product (phage cocktail), which is prepared according to the aseptic principles described in the EU Guidelines to Good Manufacturing
Practices (GMP).17 The preparation needs to be free from host-cell
proteins and DNA, pyrogenic exotoxins, endotoxins, residual agents
such as solvents, and haemolysins. For the purpose of control, the
potency of the active substance can be assessed by methods such
as pfu determination or negative-staining transmission electron
microscopy (as an orthogonal test). Participants discussed that
control of the drug product requires flexibility to accommodate
adapting the product to the diversity of bacterial strains. It was
reported that the GMP production process should support changes
in the composition of a phage preparation and that the criteria for
accepting these changes should be pre-established.
Downloaded from https://academic.oup.com/jac/article-abstract/71/8/2071/2237822 by guest on 14 June 2020
considered ATMPs, exceptions foreseen for ATMPs as defined in
Regulation (EC) number 1394/200711 and in Directive 2001/83/
EC may also apply, e.g. the possibility of a risk-based approach
to determine the extent of quality, preclinical and clinical data
required for the granting of a marketing authorization.
Workshop participants discussed various proposed approaches
in developing bacteriophage therapy.12 A ‘library’ approach entails
the creation of a phage bank from which one or several bacteriophages are selected for the individual patient, based on the characteristics of the isolated pathogen—such a concept has some
similarities with the approach for allergen products that require
a large number of substances. It has also been suggested that
phages could be cultivated and selected in the presence of the
pathogen isolated from the patient. This fully individualized concept may, from a regulatory perspective, have some similarities
with autologous cell-based products, in which an inherently variable biological material from the patient affects the final product.
However, for autologous ATMPs, the active substance (cells) will
have the same predefined characteristics (i.e. same cell type,
same cell surface markers).
The potential need for a quick change or update of a bacteriophage preparation in response to developing resistance needs
to be reconciled with the provisions of current regulations.
Changes to the terms of a marketing authorization are covered by
Commission Regulation (EC) number 1234/2008 (‘the Variations
Regulation’),13 and require time-consuming processes to ensure
that the safety and efficacy of the product is maintained. It should
be noted that changes in this context do not include the introduction
of an additional active substance (phage), because the outcome of
such an alteration would be regarded as a different medicinal product requiring a separate marketing authorization. Examples have
been brought up from the veterinary regulatory framework, where
some vaccines are allowed more flexibility for change of composition
via the inclusion of a multistrain dossier.14 However, no such exceptions currently exist within the regulatory framework for human
medicinal products. Annex I of the Variations Regulation lists
changes that require an extension of the marketing authorization
(for which the assessment follows a timeline as for initial applications). As a relevant example, replacement of a biological active
substance with one of a slightly different molecular structure,
for which the efficacy/safety characteristics are not significantly
different, is cited in Annex I of the Variations Regulation.
Precedents of exceptions do exist, as specified in the Variations
Regulation. Notably, a shorter assessment time frame is foreseen
for strain changes of the (seasonal, prepandemic or pandemic)
influenza vaccine, which are considered a major (type II) variation
rather than an extension. However, although accelerated, this
process is still a complex one, with significant time required. It
has been suggested that replacing one strain of bacteriophage
with another should follow a similar accelerated route. Further
evidence would, however, be needed to establish the data
required to ensure that the bacteriophage strain change would
not have a negative impact on the efficacy or safety of the
product.
JAC
Review
Clinical development issues
As described in the scientific literature,7,18 bacteriophage therapy
has potential advantages, including activity against Grampositive as well as Gram-negative bacteria. Importantly, bacteriophage therapy also covers MDR strains, exhibits high specificity
with narrow therapeutic spectrum (limiting disruption to the
microbial ecology) and induces few side effects so far reported
from observational studies in patients19 and in healthy volunteers.20 Nevertheless, the high specificity of bacteriophage therapy has been recognized as a potential drawback, since for
some of the proposed approaches, culturing of the causative bacteria is required prior to initiating treatment. Nevertheless, the
high specificity of bacteriophage therapy has been recognized
as a potential drawback, since for some of the proposed
approaches, culture and identification of the causative bacterium
is required prior to initiating treatment, i.e. matching bacteriophages to the infecting strains, in a time-consuming manner.
Also, a number of important issues, such as the population
dynamics of the bacteria during phage therapy, optimal duration
and modalities of treatment (topical versus systemic, stand-alone
versus concomitant use with conventional antibiotic agents),
need further exploration. Phage therapy could, however, be envisaged for many different indications, ranging from prevention of
infection (e.g. selective decontamination in the ICU) to curative
usage, e.g. treatment of skin and soft tissue infections, osteomyelitis or respiratory tract infections. Further issues to consider are
potential immune recognition with fast clearance of bacteriophages, impact on immune system, release of endotoxin upon
lysis of Gram-negative bacteria triggering a pro-inflammatory
cascading effect, as well as development of resistance by the bacterial host. With reference to the latter, the application of a cocktail of lytic bacteriophages attacking the same bacteria via
different targets appears to be a logical choice. However, whether
this approach would be the most effective has not been established. Alternatively, a sequential application of phages may be
a viable alternative, with more efficient eradication of the bacterial pathogen as resistance evolves.
There was consensus amongst participants that more robust
scientific evidence on the value of bacteriophages from adequately
designed clinical randomized studies is urgently needed. To date,
few comparative data from small-scale clinical trials with bacteriophages are available.5,6 Following a pilot study,21 a first multicentre,
open-label, randomized comparative trial is currently ongoing in
burns patients to assess tolerance and efficacy of local bacteriophage treatment of wounds infected with Escherichia coli or
Pseudomonas aeruginosa (‘Phagoburn’; ClinicalTrials.gov identifier:
NCT02116010). The primary outcome of the study is based on
microbiological criteria, measuring the time for bacteria reduction
adjusted on antibiotic treatment. Secondary outcomes involve
safety/tolerance of treatment and healing improvement. Adult
patients with burn wounds who present with local signs of infection
and with wounds showing a microbiologically documented infection (positive culture from a surface swab) with either E. coli or
P. aeruginosa, regardless of the resistance profile, are to be
included. The investigational product (phage cocktail) consists of
12 (E. coli) or 13 (P. aeruginosa) lytic phages, whilst silver sulfadiazine is designated as control treatment. These treatments are
locally applied as a daily dressing for 7 days and microbiological
swabbing is carried out twice daily during the study phase.
Patients are recruited across 11 centres using the same standardized protocol. The trial aims to recruit 220 patients over 1 year.
This trial uses a phage cocktail that is GMP compliant, and as
such various difficulties were encountered in its preparation. It
was evident during the workshop that this posed a great challenge
to the supplying manufacturer. The Phagoburn trial is to be viewed
as a ‘proof of concept’, and results will further inform subsequent
clinical research. Within the current trial, a dose–response relationship is not being investigated, and the approach towards dosefinding for bacteriophages would be significantly different from
that for conventional medicines. In future exploration, in vitro models could possibly be employed in order to properly understand the
treatment dynamics. Other issues, such as impact on gut microbiota including the important concern of potential transmission
of resistance genes; modulation of immune responses; bacterial
replacement with ensuing secondary infections; and ultimate positioning of lytic bacteriophages as part of the therapeutic armamentarium need due consideration.
To ensure successful development of bacteriophage therapy so
as to reach patients in need at the earliest, a regular dialogue with
regulatory authorities is warranted. Drug developers should take
advantage of early exchanges with regulators and of subsequent
formal assistance offered by the scientific-advice process.22 The
latter offers a platform for advice on questions concerning quality,
non-clinical and clinical aspects of the drug development plan, as
well as on issues relating to interpretation and implementation
of (draft) EU guidelines; this process ultimately increases the
probability of a positive regulatory outcome.23
Conclusions
Bacteriophage therapy is a somewhat unique approach to address
the need for alternatives to antibacterial medicines in an era of bacterial multidrug resistance. However, there are regulatory and scientific challenges with phage therapy. The EMA workshop, which
was attended by various stakeholders, explored the bottlenecks
hindering successful therapeutic innovation with bacteriophages.
It was clear from this workshop that there are several manufacturers progressing with the development of investigational
bacteriophage cocktails to be tested in clinical trials. It was also
2073
Downloaded from https://academic.oup.com/jac/article-abstract/71/8/2071/2237822 by guest on 14 June 2020
Participants acknowledged that bacteriophage therapy, which
by its very nature involves a co-evolutionary dynamic pattern
between bacteria and lytic phages, poses a unique challenge for
regulators. Directive 2001/83/EC10 states that an application for a
marketing authorization should be accompanied by qualitative
and quantitative particulars of all the constituents of the medicinal
product. Biological active substances have inherent variability;
nevertheless, they must be appropriately characterized, including
identifying critical parameters relevant to produce a high-quality,
well-characterized and safe product. Specifications and appropriate acceptance criteria also need to be set and justified, and the
manufacturing process should be adequately described, validated
and controlled in order to ensure that the product is of consistent
quality. Bacteriophage therapy should adhere to these prerequisites; however, the scientific requirements and necessary flexibility require further discussion and definition to accommodate the
need for rapid change in composition of the phage preparation.
Review
Funding
The workshop was supported by EMA institutional funding only.
Transparency declarations
None to declare.
Disclaimer
The views expressed in this paper are the personal views of the authors
and must not be understood or quoted as being made on behalf of or
representing the position of the EMA or one of its committees or working
parties.
References
1 Twort FW. An investigation on the nature of ultra-microscopic viruses.
Lancet 1915; 189: 1241– 3.
2 d’Hérelle F. Sur un microbe invisible antagoniste des bacilles dysentériques. C R Acad Sci Paris 1917; 165: 373–5.
3 Bruynoghe R, Maisin J. Essais de thérapeutique au moyen du bacteriophage du Staphylocoque. Compt Rend Soc Biol 1921; 85: 1120– 1.
4 Sulakvelidze A, Alavidze Z, Morris JG Jr. Bacteriophage therapy.
Antimicrob Agents Chemother 2001; 45: 649–59.
5 Rhoads DD, Wolcott RD, Kuskowski MA et al. Bacteriophage therapy of
venous leg ulcers in humans: results of a phase I safety trial. J Wound
Care 2009; 18: 237–8, 240– 3.
6 Wright A, Hawkins CH, Anggård EE et al. A controlled clinical trial of a
therapeutic bacteriophage preparation in chronic otitis due to antibiotic
resistant Pseudomonas aeruginosa; a preliminary report of efficacy. Clin
Otolaryngol 2009; 34: 349– 57.
7 Wittebole X, De Roock S, Opal SM. A historical overview of bacteriophage
therapy as an alternative to antibiotics for the treatment of bacterial
pathogens. Virulence 2014; 5: 226– 35.
8 Verbeken G, Pirnay JP, De Vos D et al. Optimising the European regulatory
framework for sustainable bacteriophage therapy in human medicine.
Arch Immunol Ther Exp 2012; 60: 161– 72.
Pharmaceutical Legislation: Medicinal Products For Human Use. European
Commission. http://ec.europa.eu/health/files/eudralex/vol-1/dir_2001_
83_consol_2012/dir_2001_83_cons_2012_en.pdf.
11 European Commission. Regulation (EC) no 1394/2007 of the European
Parliament and of the council of 13 November 2007 on advanced therapy
medicinal products and amending directive 2001/83/EC and regulation
(EC) no 726/2004 (consolidated version: 02/07/2012). In: EudraLex—The
Rules Governing Medicinal Products in the European Union, Volume 1:
Pharmaceutical Legislation: Medicinal Products For Human Use. European
Commission. http://ec.europa.eu/health/files/eudralex/vol-1/reg_2007_
1394/reg_2007_1394_en.pdf.
12 Chan BK, Abedon ST, Loc-Carrillo C. Phage cocktails and the future of
phage therapy. Future Microbiol 2013; 8: 769– 83.
13 European Commission. Regulation (EC) no 1234/2008 concerning the
examination of variations to the terms of marketing authorisations for
medicinal products for human use and veterinary medicinal products
(consolidated version: 02/11/2012). In: EudraLex—The Rules Governing
Medicinal Products in the European Union, Volume 1: Pharmaceutical
Legislation: Medicinal Products For Human Use. European Commission.
http://ec.europa.eu/health/files/eudralex/vol-1/reg_2008_1234_cons_
2012-11-02/reg_2008_1234_cons_2012-11-02_en.pdf.
14 Committee for Medicinal Products for Veterinary Use. Guideline on Data
Requirements for Multi-strain Dossiers for Inactivated Vaccines against
Avian Influenza (AI), Bluetongue (BT) and Foot-and-Mouth Disease (FMD).
European Medicines Agency. http://www.ema.europa.eu/ema/pages/
includes/document/open_document.jsp?webContentId=WC500077950.
15 International Conference on Harmonisation of Technical Requirements
for Registration of Pharmaceuticals for Human Use, Committee for
Proprietary Medicinal Products. CHMP/ICH Q5D Note for Guidance on
Quality of Biotechnology Products: Derivation and Characterisation of Cell
Substrates Used for Production of Biotechnological/Biological Products
(CPMP/ICH/294/95). European Medicines Agency. http://www.ema.
europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/
WC500003280.pdf.
16 Pirnay JP, Blasdel BG, Bretaudeau L et al. Quality and safety requirements for sustainable phage therapy products. Pharm Res 2015; 32:
2173– 9.
17 European Commission. Annex 1: manufacture of sterile medicinal products. In: EudraLex—The Rules Governing Medicinal Products in the
European Union, Volume 4: EU Guidelines to Good Manufacturing Practice,
Medicinal Products for Human and Veterinary Use. European Commission.
http://www.ec.europa.eu/health/files/eudralex/vol-4/2008_11_25_gmpan1_en.pdf.
18 Parracho HMRT, Burrowes BH, Enright MC et al. The role of regulated
clinical trials in the development of bacteriophage therapeutics. J Mol
Genet Med 2012; 6: 279–86.
19 Mie˛dzybrodzki R, Borysowski J, Weber-Da˛browska B et al. Clinical
aspects of phage therapy. Adv Virus Res 2012; 83: 73 –121.
20 Bruttin A, Brüssow H. Human volunteers receiving Escherichia coli
phage T4 orally: a safety test for phage therapy. Antimicrobiol Agents
Chemother 2005; 49: 2558– 69.
21 Rose T, Verbeken G, de Vos D et al. Experimental phage therapy of burn
wound infection: difficult first steps. Int J Burns Trauma 2014; 4: 66 –73.
9 Verbeken G, Pirnay JP, Lavigne R et al. Call for a dedicated European legal
framework for bacteriophage therapy. Arch Immunol Ther Exp 2014; 62:
117–29.
22 European Medicines Agency. Scientific Advice and Protocol Assistance.
http://www.ema.europa.eu/ema/index.jsp?curl=pages/regulation/general/
general_content_000049.jsp&mid=WC0b01ac05800229b9.
10 Directive 2001/83/EC of the European Parliament and of the council of
6 November 2001 on the community code relating to medicinal products
for human use (consolidated version: 16/11/2012). In: EudraLex—The
Rules Governing Medicinal Products in the European Union, Volume 1:
23 Regnstrom J, Koenig F, Aronsson B et al. Factors associated with
success of market authorisation applications for pharmaceutical drugs
submitted to the European Medicines Agency. Eur J Clin Pharmacol
2010; 66: 39– 48.
2074
Downloaded from https://academic.oup.com/jac/article-abstract/71/8/2071/2237822 by guest on 14 June 2020
evident that physicians within the EU are already treating patients
with this therapy on a compassionate use basis. However, there is
currently a considerable lack of robust evidence to prove the efficacy and safety of these products. More data are urgently needed
on the subject; it is anticipated that ongoing research efforts will
inform follow-up research and debate. An ongoing dialogue
between the drug developers and regulatory authorities on how
the regulatory framework might support and offer appropriate
flexibility in delineating the tests and studies to be undertaken is
highly encouraged. This approach would facilitate the introduction of bacteriophage therapy in clinical practice if such therapy
is proven to be safe and effective.