Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
Published OnlineFirst March 27, 2013; DOI: 10.1158/1078-0432.CCR-12-3661 Infection and Cancer: Revaluation of the Hygiene Hypothesis Katerina Oikonomopoulou, Davor Brinc, Kyriacos Kyriacou, et al. Clin Cancer Res Published OnlineFirst March 27, 2013. Updated version E-mail alerts Reprints and Subscriptions Permissions Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-12-3661 Sign up to receive free email-alerts related to this article or journal. To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at pubs@aacr.org. To request permission to re-use all or part of this article, contact the AACR Publications Department at permissions@aacr.org. Downloaded from clincancerres.aacrjournals.org on May 20, 2013. © 2013 American Association for Cancer Research. Published OnlineFirst March 27, 2013; DOI: 10.1158/1078-0432.CCR-12-3661 Clinical Cancer Research Review Infection and Cancer: Revaluation of the Hygiene Hypothesis Katerina Oikonomopoulou1, Davor Brinc2, Kyriacos Kyriacou3, and Eleftherios P. Diamandis1,2 Abstract Several studies have shown that persistent infections and inflammation can favor carcinogenesis. At the same time, certain types of pathogens and antitumor immune responses can decrease the risk of tumorigenesis or lead to cancer regression. Infectious agents and their products can orchestrate a wide range of host immune responses, through which they may positively or negatively modulate cancer development and/or progression. The factors that direct this dichotomous influence of infection-mediated immunity on carcinogenesis are not well understood. Even though not universal, several previous reports have investigated the inverse link of pathogen-induced "benign" inflammation to carcinogenesis and various other pathologies, ranging from autoimmune diseases to allergy and cancer. Several models and ideas are discussed in this review, including the impact of decreased exposure to pathogens, as well as the influence of pathogen load, the timing of infection, and the type of instigated immune response on carcinogenesis. These phenomena should guide future investigations into identifying novel targets within the microbial and host proteome, which will assist in the development of cancer therapeutics and vaccine remedies, analogous to earlier efforts based on helminthic components for the prevention and/or treatment of several pathologies. Clin Cancer Res; 19(11); 1–8. 2013 AACR. Introduction Even though the course of carcinogenesis is undoubtedly multifactorial, major attention has been attracted on the role of infectious diseases and the immune system in cancer development (1–4). Several types of carcinomas are related to infections (2, 5), whereas inflammation is recognized as one of the hallmarks of cancer (1, 6), and inclusion of immunologic assessments in cancer classification and prognosis has been suggested (7, 8). In contrast, immune responses, including those triggered by microorganisms, are known to decrease cancer risk or lead to tumor regression. The relationship between infection and tumorigenesis is not well understood, and both favorable and unfavorable immune-mediated or direct anticarcinogenic microbial effects have been observed. This review aims to provide an update primarily on the inverse association between infections and cancer and provide clues for potential underlying mechanisms. Attention is drawn to the hygiene hypothesis that attempts to explain the increased incidence of pathol- Authors' Affiliations: 1Department of Pathology and Laboratory Medicine, Mount Sinai Hospital; 2Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada; and 3Department of EM/Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus K. Oikonomopoulou and D. Brinc contributed equally to this work. Corresponding Author: Eleftherios P. Diamandis, Department of Pathology & Laboratory Medicine, Mount Sinai Hospital, 60 Murray Street, Room L6-201, Toronto, ON M5T 3L9. Phone: 416-586-4800, ext. 8813; Fax: 416619-5521; E-mail: ediamandis@mtsinai.on.ca doi: 10.1158/1078-0432.CCR-12-3661 2013 American Association for Cancer Research. ogies such as allergies, autoimmune diseases, and cancer in the industrial world. Several historical observations and other theories, such as hormesis (9) and concomitant immunity (10), are revisited to lend more credence to the hygiene hypothesis. The Cancer Hygiene Hypothesis Several decades ago, the hygiene hypothesis, referring to the lack of exposure to microbes at childhood, was introduced to explain the higher numbers of allergic and autoimmune diseases in the Western world and urbanized communities (11–14). More recently, the hygiene hypothesis has been restated to account for the association between microorganisms and cancer (13). Following the same pattern observed with some immune pathologies, there is growing evidence of an increased cancer incidence in Westernized economically developed countries (15). Socioeconomic status was also inversely associated with Hodgkin lymphoma (16), and daycare attendance was associated with a lower risk of acute lymphoblastic leukemia (17, 18). The resemblance of the hygiene–immunopathology relationship to the one exhibited by hygiene and cancer is not surprising, given that preliminary observations have associated tumorigenesis with chronic immune-mediated disorders (Table 1); for example, an increased risk of cancer has been observed in patients with autoimmune disease (19, 20), chronic allergic disorders have been connected to pro- and antitumor effects (21–24), and allergic patients with cancer have been suggested to exhibit higher cure rates and more favorable disease progression (25). Some experimental evidence may also support the cancer hygiene hypothesis, that is, the antitumorigenic role of several www.aacrjournals.org Downloaded from clincancerres.aacrjournals.org on May 20, 2013. © 2013 American Association for Cancer Research. OF1 Published OnlineFirst March 27, 2013; DOI: 10.1158/1078-0432.CCR-12-3661 Oikonomopoulou et al. Table 1. Association between different pathologies and cancer, based on epidemiologic and experimental studies Condition Infections Helminths and protozoa Viruses Bacteria Allergy Autoimmune diseases Association with cancer References Negativea (13, 58, 80, 93–95) Positiveb Negativea Positiveb Negativea Positiveb Negativea Positiveb Positiveb (43, 96, 97) (57) (2, 5, 13, 98) (13, 26, 28, 47, 54–56) (2, 5, 13, 27, 37, 98) (21–24) (21, 24) (19, 20) a Negative: cancer prevention, cancer regression, decreased cancer risk. b Positive: cancer promotion, increased cancer risk. inflammatory components, the ability of some commensals and benign gastrointestinal parasites like helminths to downregulate inflammation, as well as the ability of pathogens and their products to stimulate anticancer immunity (see sections below). However, the hygiene hypothesis, as it stands, cannot rationalize why specific infectious agents (e.g., Helicobacter pylori; refs. 26, 27) or microbial products [e.g., lipopolysaccharide (LPS); refs. 28, 29] can exhibit both pro- and anticarcinogenic functions and, therefore, many questions remain unresolved. Immune Responses to Infection and Cancer Host immune response to pathogens generally involves effectors preexisting locally in mucus (e.g., immunoglobulin A, antimicrobial peptides, lysozyme) or plasma (natural immunoglobulin M, complement), followed by activation of more specialized innate (e.g., macrophages, granulocytes, dendritic, mast, natural killer cells) and adaptive (T cells, B cells) immune processes, to facilitate clearance of pathogens or reduction of their impact (30). Innate immune cell activation can trigger phagocytosis, release of antimicrobial compounds and proinflammatory cytokines, as well as lead to immune suppression, fibrosis, angiogenesis, and wound healing (31). T cells, following pathogen recognition and depending on the antigen and local environment, develop into CTL or Thelper cells (TH), namely TH1, TH17, or TH2 cells, mediating different cytokine expression patterns, known as classical (TH1, TH17) or alternative (TH2) inflammation (32). TH cells also stimulate production of antibodies from antigen-activated B cells. Another distinct cell subtype, regulatory T cells (Treg), particularly observed in chronic parasitic infections (e.g., helminths), have a role OF2 Clin Cancer Res; 19(11) June 1, 2013 in preventing immune-mediated damage (33, 34). Notably, the immune response pattern can vary during the infection course; in helminth infections, a TH1 to TH2 shift is commonly observed in parallel with infection progression, and may also signal the reduced effectiveness of a drug therapy (35, 36). The various immune processes induced during infection may also be implicated in cancer. In 1863, it was Rudolf Virchow who showed the presence of leukocytes in neoplastic tissue (reviewed in ref. 37). Paul Ehrlich later suggested that the immune system continuously destroys spontaneously arising tumors (immune surveillance hypothesis), work that was updated by the cancer immunoediting hypothesis, stating that the immune system has a significant role in shaping the properties of an emerging tumor (38, 39). Both innate and adaptive immune cells are now known to localize at tumor sites, with specific cell subsets, densities, and intratumor locations being associated with cancer risk or survival (8). Antibodies against tumor-associated antigens have also been detected in cancer patients’ sera (International SEREX Program, The Ludwig Institute for Cancer Research, Uppsala, Sweden). However, although several studies have considered the role of immunity in cancer survival/progression, the idea that an existing infection may further modulate the pro-/antitumorigenic immune effect has been overlooked. Infection as a Carcinogenic Factor Some infectious agents can directly influence carcinogenesis; for instance, human papillomavirus protein E7 can bind the retinoblastoma tumor suppressor and the cyclindependent kinase inhibitor p21 in infected cells, promoting DNA replication and cell proliferation (40), whereas Hepatitis B virus can induce hypoxia-inducible factor-1a, stimulating angiogenesis (41). Pathogens may also promote tumorigenesis indirectly (Table 1; refs. 3, 4), by activating cancer-mediating host inflammatory pathways. The helminth Schistosoma haematobium can induce urothelial dysplasia and inflammation upon intravesical administration in mice (42) and has been linked to bladder cancer (43). In another example, Propionibacterium acnes, found in prostate cancer and benign hyperplasia samples, when cocultured with prostate epithelial cells results in production of proinflammatory cytokines, prostaglandins, and activated matrix metalloproteinases, whereas long-term infection leads to anchorage-independent cancer cell growth (44). Inflammation induced by chronic infections may be able to trigger mutations, epigenetic changes, and protein modifications that may lead to oncogene activation and tumor suppressor inhibition (3). Apart from the typical infectious agents, altered intestinal microbiota may also promote carcinogenesis, DNA damage, and cell proliferation via chronic inflammatory processes (45). Secretion of pathogen-induced cytokines may also have a dual role depending on the settings; for example, TNFa can mediate tumor hemorrhagic necrosis and regression (46, 47), whereas, on the other hand, it can promote carcinogenesis if present in a chronic fashion (48). Clinical Cancer Research Downloaded from clincancerres.aacrjournals.org on May 20, 2013. © 2013 American Association for Cancer Research. Published OnlineFirst March 27, 2013; DOI: 10.1158/1078-0432.CCR-12-3661 Infection and Cancer In addition to giving rise to the inflammation-mediated detrimental effects, pathogens may also promote tumorigenesis by inhibiting host anticancer immunity, for instance, by stimulating production of immunosuppressive cytokines [e.g., interleukin (IL)-10], causing T-cell apoptosis, promoting T-cell subtypes with attenuated antitumor activity (e.g., TH2), or triggering recruitment of myeloid suppressor cells and Tregs (49–52). Another potential effect on antitumor immunity triggered by chronic infections, also observed in cancer, is the dysfunction and subsequent elimination of antigen-specific T cells, a phenomenon called T-cell exhaustion (53). Infection in Cancer Prevention Several observations, reported as early as the 1700s, support the link between infection-mediated inflammation and cancer prevention or regression (Table 1); most notable are the efforts by William Coley in early 19th century to vaccinate his patients with cancer with an attenuated bacterial mixture (Streptococcus pyogenes and Serratia marcescens) that accomplished significant cure and favorable progression rates (47, 54). There is also evidence of the antitumor effect of certain microbial products (e.g., LPS) and attenuated pathogen forms [e.g., Bacillus Calmette-Guerin (BCG) vaccine; refs. 13, 28, 55]; more specifically, BCG, vaccinia, or yellow fever virus vaccinations have been linked to melanoma protection (56, 57). In addition, infectious agents have also been inversely associated with cancer (Table 1), as in the case of Trypanosoma cruzi, which can result in lower incidence of experimentally induced rodent colon cancer (58). These observations support the protective action of infections, as proposed by the hygiene hypothesis (11–14). In the subsequent sections, we will expand on this discussion by suggesting potential mechanisms that are often underestimated but may likely explain the favorable association of infection to carcinogenesis (Fig. 1). Inlammation/Immunity Suppression Activation Antigen cross-reactivity Angiogenesis inhibition Immune cells STOP STOP Pathogens Carcinogen removal Tumor cells Preexisting immunity Low-dose High-dose effects effects Pathogen-induced antitumor mechanisms Hormetic effects Tumor microenvironment Threshold © 2013 American Association for Cancer Research Figure 1. Potential pathogen-mediated antitumor mechanisms. A microorganism may influence the fine balance between immunosuppression and immunity against a concurrent or subsequent tumor by modulating the availability and presentation of cross-reactive antigens, by influencing induction of preexisting immunity, and by shaping the components of the tumor microenvironment. The levels of microbe-triggered stimuli are also decisive factors on the biphasic influence (pro- or anti-inflammatory) that a microorganism can have on immune functions. Several other mechanisms, such as removal of carcinogens and restriction of tumor vascularization may also facilitate the beneficial antitumor effects of microbes on their host. www.aacrjournals.org Clin Cancer Res; 19(11) June 1, 2013 Downloaded from clincancerres.aacrjournals.org on May 20, 2013. © 2013 American Association for Cancer Research. OF3 Published OnlineFirst March 27, 2013; DOI: 10.1158/1078-0432.CCR-12-3661 Oikonomopoulou et al. Suppression of inflammation Several microbial products (e.g., lysophosphatidylserine) can have anti-inflammatory effects (34, 59); for instance, they can suppress toll-like receptor signaling, inflammatory cytokine and nitric oxide production, as well as inhibit innate immune cell activation and stimulate production of immunosuppressive cytokines and recruitment of Tregs. In this regard, TH1 or TH2 responses to some helminth infections rarely result in severe pathology (36) and can, in fact, downregulate allergic or autoimmune pathology (e.g., see ref. 60). However, immunosuppressive cytokines, specifically, have pleiotropic effects on tumorigenesis either by inhibiting inflammation-associated tumorigenesis or by restricting antitumor immunity; for example, IL-10 is known to either inhibit or promote tumor growth, as well as facilitate tumor rejection in mice (61). Promotion of antitumor immunity Microorganisms may provide specific triggers (e.g., lowlevel endotoxin, commonly produced by many pathogens) that increase antigenicity of nascent tumor cells, or keep the immune cells in an "alerted" immunosurveillance state (13). This phenomenon resembles the infection-mediated stimulation of autoimmunity as a result of molecular mimicry, epitope spreading, exposure of cryptic antigens, or bystander activation (62). Epitope spreading has been observed in few cancer vaccine studies, i.e., following injection of dendritic cells in patients with melanoma (63). An infection can also lead to tumor cell destruction, subsequent release of tumor antigens, and activation of antigen-presenting cells. This could potentially trigger T-cell responses with antitumor activities, like the ones that may be responsible for the protective action of BCG (64). Moreover, potential increases in tumor vascular permeability may also facilitate the local recruitment of anticancer T cells (47). Heat shock proteins expressed by stressed cells and found upregulated in virus-infected and several cancer cells can also be immunogenic, thus influencing antitumor responses (65). Presentation of cross-reactive antigens Several pathogens contain antigens, mainly glycoproteins, that cross-react with tumor-associated antigens. As an example of such glycoprotein cross-reactivity, the Thomsen–Friedenreich T and Tn parasitic antigens can be detected in more than 80% of patients with cancer and have been under experimental and clinical investigations as markers and therapeutic targets for cancer (66, 67). Furthermore, sera from patients suffering from parasitic infections (e.g., Echinococcus) are commonly found to crossreact (contain similar immunogenic epitopes) with sera from patients with cancer (68). Interestingly, it has been observed that such sera are more frequent in patients with less extensive malignancy. Antibodies against these shared parasite/tumor-associated antigens can potentially target tumor cells for destruction or promote antigen presentation to T cells and induce antitumor responses; this antibodymediated immune enhancement has been observed for nontumor antigens in experimental models (69). OF4 Clin Cancer Res; 19(11) June 1, 2013 Induction of preimmunity The "concomitant immunity hypothesis" was originally suggested to explain resistance to secondary tumors or infections, particularly in animal models (10, 70). As an ongoing persistent infection can protect the host from the same infection, similarly, in animal models, immunity to the original tumor can prevent growth of a comparable mass (10, 71). Concomitant immunity was considered the result of either immunogenic factors, for example common antigenic epitopes, or nonimmunogenic factors, such as putative antimitotic components (10). The concomitant effect may be abrogated once the original tumor is removed. It has also been observed that anticancer immunity can be present after the removal of the original malignant mass, a phenomenon termed sinecomitant immunity (10, 71) that can potentially be attributed to the parallel removal of tumor-induced immunosuppression. Formulation of the tumor microenvironment In principle, any agent that modulates antigen expression and cell populations in the tumor microenvironment can determine the quality and level of anticancer immunity. For instance, the previously observed effect of Coley’s toxin on cancer regression may be the result of TNFa affecting local vascular permeability and enhancing leukocyte recruitment (47, 54). Microorganisms, such as helminthes and commensals, may also contribute to a cancer inhibitory microenvironment by affecting TH1/TH2 responses and Tregs recruitment (36, 72). Infection-mediated antitumor immunity can also be restricted by the immunosuppressive microenvironment that is often associated with developed tumors and characterized by TH2 responses and the presence of myeloid-derived suppressor cells and Tregs (73). Tumorassociated macrophages can also promote angiogenesis, tumor cell invasion, metastasis, and T-cell inhibition. Angiogenesis itself has been related to immune suppression; for example, VEGF may lead to decreased antigen presentation to T cells, due to inhibition of dendritic cells maturation (74). The role of microbial infections in forming the local versus systemic or "secondary" (noninfected site) pro- or anticarcinogenic immune milieu in competition with the immunosuppressive tumor microenvironment remains to be discovered. Production of low-level "danger" signals A phenomenon termed hormesis has been coined to describe a biphasic dose-dependent response to an agent characterized by a low-dose beneficial effect and a highdose inhibitory or toxic effect (9). It can be speculated that microbes, and specifically relatively benign microorganisms and commensals, embody this pleiotropic response by stimulating DNA and tissue repair processes at low infectious agent loads while resulting in extensive inflammatory and genomic changes that can subsequently foster procarcinogenic processes at higher pathogen loads. Interestingly, it has been postulated that the hygiene hypothesis describes this beneficial low-level exposure phenomenon (75). As the hormetic effect would Clinical Cancer Research Downloaded from clincancerres.aacrjournals.org on May 20, 2013. © 2013 American Association for Cancer Research. Published OnlineFirst March 27, 2013; DOI: 10.1158/1078-0432.CCR-12-3661 Infection and Cancer be highly dependent on spatial and temporal factors, in the case of carcinogenesis, both tumor stage and location at the time of infection may be of paramount importance; the beneficial effects of pathogen-triggered stress repair processes at tumor initiation may, therefore, be replaced by detrimental effects in later stages, when repair may be accompanied by a more immunosuppressive microenvironment. Removal of carcinogens The health benefits of bacterially enriched food (probiotics) and certain ingredients that can stimulate growth of indigenous commensal bacteria (prebiotics) have been widely discussed in several settings, including inflammation (76). Although the evidence for the ability of probiotics to reduce risk of colorectal cancer is still controversial, probiotics, particularly containing bifidobacteria and lactobacilli, have been suggested to reduce the production of carcinogens by other gastrointestinal bacteria like clostridia and bacteroides (77). Inhibition of angiogenesis It has been suggested that infection can prevent angiogenesis, an effect that may subsequently lead to restriction of tumor growth. For example, despite its potential role in induction of tumor-promoting myeloid suppressor cells (78, 79), Toxoplasma gondii infection is also known to suppress vascularization in a mouse melanoma model, an effect that may be attributed in part to secretion of antiangiogenic cytokines (80). Cancer Immunotherapy and Pathogen-Based Therapeutics The concept of using anti-inflammatory agents to regulate not only immune processes but also the tumor load is not new, with the most widely discussed recent example being the benefits of aspirin in carcinogenesis risk reduction (81). The latest approach in immune-related cancer therapy is to promote targeting of specific tumor antigens or stimulate the host immune response to growing tumors using a number of different approaches (82, 83). Several tumor cell antigens, that is, cancer specific, differentiation, viral, and carbohydrate, as well as mutated and overexpressed proteins have been considered as potential vaccine candidates (e.g., see ref. 84). In addition, antibody-based therapeutic agents with reduced immunogenicity have been designed to specifically recognize and destroy tumor cells directly or via their specific stromal or immunomodulatory effects (82). T cells have also been investigated in cancer treatment, for example, in patients with leukemia and melanoma (85). In addition, Tregs from mice infected by selected pathogens (e.g., Helicobacter hepaticus) have exhibited anticancer activity (86). In a more microbe-based approach, pathogens and their toxins have been tested as antitumor agents or as carriers for tumor-targeting therapies (87). The concept behind this approach is to use the infectious agent or www.aacrjournals.org its selected components as means to treat/prevent cancer. In this regard, the BCG vaccine, an attenuated form of Mycobacterium bovis, is now an U.S. Food and Drug Administration–approved agent for the first-line intravesical treatment of bladder cancer (55). BCG in this context may have a role in stimulating the body’s own anticancer immunity via enhancing TH1 cytokine production (e.g., IFN-g, TNFa; refs. 88, 89). Microbial components may also find applicability in preventing cancer, as in the case of the tumor-pathogen T/Tn antigen (90) and the bacterial endotoxin LPS (28). More specifically for the T/Tn antigen, vaccination regimens based on this common microbe–tumor glycoprotein (66, 67) have been previously evaluated in breast cancer prevention (90). Vaccination was accompanied by an increase of helper T lymphocytes and decrease of T-suppressor/cytotoxic cell ratio, possibly leading to regulation of antitumor immune responses and subsequent prevention of breast cancer recurrence. More recently, the helminth Trichuris suis has been under clinical and experimental investigation for its ability to alleviate diseases, such as inflammatory bowel disease (ulcerative colitis, Crohn disease), multiple sclerosis, and allergy (e.g., see ref. 91, 92). Its applicability to cancer pathology, and more specifically to tumors of the gastrointestinal system, is a question open to future investigations. Conclusion and Future Perspectives Both protective and detrimental effects of microorganisms have been observed, many of them linked to various immune components. Overall, their effect may depend on the fine orchestration between induction and suppression of cancer-promoting or antitumorigenic immunity as well as on the level of pathogen load and the timing between infection and cancer initiation. In this regard, cancer may be associated with the increased hygiene/ decreased exposure to specific microorganisms, similar to what is known for autoimmune diseases and allergies. That said, it should be noted that not all types of microorganisms are expected to have the same anticarcinogenic effect; for example, viral infections seem to be mainly procarcinogenic, in contrast to bacteria or parasitic worms that have a longer coevolution history with human species and may have, therefore, adapted to exhibit more antitumorigenic effects. Novel clinical studies are therefore needed to delineate the specific role of these relatively benign organisms in modulating the host immune response toward cancer prevention. The adjuvant and cross-reactive effects of parasites and commensals should be investigated in more detail to identify potential novel therapeutic targets. Exploration of the immunogenic epitope availability orchestrated by these agents may also, in the future, assist in the development of personalized treatments and immunization strategies that can be used to prevent, regress, or slow down cancer progression. Clin Cancer Res; 19(11) June 1, 2013 Downloaded from clincancerres.aacrjournals.org on May 20, 2013. © 2013 American Association for Cancer Research. OF5 Published OnlineFirst March 27, 2013; DOI: 10.1158/1078-0432.CCR-12-3661 Oikonomopoulou et al. Disclosure of Potential Conflicts of Interest No potential conflicts of interest were disclosed. Authors' Contributions Conception and design: K. Oikonomopoulou, D. Brinc, K. Kyriacou, E.P. Diamandis Writing, review, and/or revision of the manuscript: K. Oikonomopoulou, D. Brinc, K. Kyriacou, E.P. Diamandis Study supervision: K. Oikonomopoulou, K. Kyriacou, E.P. Diamandis Acknowledgments The authors apologize for not citing more reviews and original papers related to this topic due to space limitations. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Received November 28, 2012; revised March 20, 2013; accepted March 21, 2013; published OnlineFirst March 27, 2013. References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. OF6 Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 2009;30:1073–81. de Martel C, Franceschi S. Infections and cancer: established associations and new hypotheses. Crit Rev Oncol Hematol 2009;70: 183–94. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010;140:883–99. Trinchieri G. Cancer and inflammation: an old intuition with rapidly evolving new concepts. Annu Rev Immunol 2012;30:677–706. Herrera LA, Benitez-Bribiesca L, Mohar A, Ostrosky-Wegman P. Role of infectious diseases in human carcinogenesis. Environ Mol Mutagen 2005;45:284–303. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646–74. Galon J, Pages F, Marincola FM, Angell HK, Thurin M, Lugli A, et al. Cancer classification using the Immunoscore: a worldwide task force. J Transl Med 2012;10:205. Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012;12:298–306. Mattson MP. Hormesis defined. Ageing Res Rev 2008;7:1–7. Gorelik E, Segal S, Feldman M. On the mechanism of tumor "concomitant immunity". Int J Cancer 1981;27:847–56. Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med 2002;347:911–20. Strachan DP. Family size, infection and atopy: the first decade of the "hygiene hypothesis". Thorax 2000;55 Suppl 1:S2–10. Rook GA, Dalgleish A. Infection, immunoregulation, and cancer. Immunol Rev 2011;240:141–59. Ngoi SM, Sylvester FA, Vella AT. The role of microbial byproducts in protection against immunological disorders and the hygiene hypothesis. Discov Med 2011;12:405–12. Rastogi T, Devesa S, Mangtani P, Mathew A, Cooper N, Kao R, et al. Cancer incidence rates among South Asians in four geographic regions: India, Singapore, UK and US. Int J Epidemiol 2008;37:147–60. Gutensohn N, Cole P. Childhood social environment and Hodgkin's disease. N Engl J Med 1981;304:135–40. Ma X, Buffler PA, Selvin S, Matthay KK, Wiencke JK, Wiemels JL, et al. Daycare attendance and risk of childhood acute lymphoblastic leukaemia. Br J Cancer 2002;86:1419–24. Gilham C, Peto J, Simpson J, Roman E, Eden TO, Greaves MF, et al. Day care in infancy and risk of childhood acute lymphoblastic leukaemia: findings from UK case-control study. BMJ 2005;330:1294. Hemminki K, Liu X, Ji J, Sundquist J, Sundquist K. Autoimmune disease and subsequent digestive tract cancer by histology. Ann Oncol 2011;23:927–33. Hemminki K, Liu X, Ji J, Sundquist K, Sundquist J. Subsequent COPD and lung cancer in patients with autoimmune disease. Eur Respir J 2011;37:463–5. Merrill RM, Isakson RT, Beck RE. The association between allergies and cancer: what is currently known? Ann Allergy Asthma Immunol 2007;99:102–16. Maisonneuve P, Lowenfels AB, Bueno-de-Mesquita HB, Ghadirian P, Baghurst PA, Zatonski WA, et al. Past medical history and pancreatic cancer risk: Results from a multicenter case-control study. Ann Epidemiol 2010;20:92–8. Clin Cancer Res; 19(11) June 1, 2013 23. Prizment AE, Folsom AR, Cerhan JR, Flood A, Ross JA, Anderson KE. History of allergy and reduced incidence of colorectal cancer, Iowa Women's Health Study. Cancer Epidemiol Biomarkers Prev 2007;16: 2357–62. 24. Engkilde K, Thyssen JP, Menne T, Johansen JD. Association between cancer and contact allergy: a linkage study. BMJ Open 2011;1:e000084. 25. Pompei R, Lampis G, Ingianni A, Nonnis D, Ionta MT, Massidda B. Allergy and tumour outcome after primary cancer therapy. Int Arch Allergy Immunol 2004;133:174–8. 26. Vaezi MF, Falk GW, Peek RM, Vicari JJ, Goldblum JR, Perez-Perez GI, et al. CagA-positive strains of Helicobacter pylori may protect against Barrett's esophagus. Am J Gastroenterol 2000;95:2206–11. 27. Kim SS, Ruiz VE, Carroll JD, Moss SF. Helicobacter pylori in the pathogenesis of gastric cancer and gastric lymphoma. Cancer Lett 2011;305:228–38. 28. Lundin JI, Checkoway H. Endotoxin and cancer. Environ Health Perspect 2009;117:1344–50. 29. Ikebe M, Kitaura Y, Nakamura M, Tanaka H, Yamasaki A, Nagai S, et al. Lipopolysaccharide (LPS) increases the invasive ability of pancreatic cancer cells through the TLR4/MyD88 signaling pathway. J Surg Oncol 2009;100:725–31. 30. Ayres JS, Schneider DS. Tolerance of infections. Annu Rev Immunol 2012;30:271–94. 31. Mosser DM. The many faces of macrophage activation. J Leukoc Biol 2003;73:209–12. 32. Zhou L, Chong MM, Littman DR. Plasticity of CD4þ T cell lineage differentiation. Immunity 2009;30:646–55. 33. Roncarolo MG, Battaglia M. Regulatory T-cell immunotherapy for tolerance to self antigens and alloantigens in humans. Nat Rev Immunol 2007;7:585–98. 34. van Riet E, Hartgers FC, Yazdanbakhsh M. Chronic helminth infections induce immunomodulation: consequences and mechanisms. Immunobiology 2007;212:475–90. 35. Mourglia-Ettlin G, Marques JM, Chabalgoity JA, Dematteis S. Early peritoneal immune response during Echinococcus granulosus establishment displays a biphasic behavior. PLoS Negl Trop Dis 2011;5: e1293. 36. Zhang W, Ross AG, McManus DP. Mechanisms of immunity in hydatid disease: implications for vaccine development. J Immunol 2008;181: 6679–85. 37. Macarthur M, Hold GL, El-Omar EM. Inflammation and cancer II. Role of chronic inflammation and cytokine gene polymorphisms in the pathogenesis of gastrointestinal malignancy. Am J Physiol Gastrointest Liver Physiol 2004;286:G515–G520. 38. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 2002;3:991–8. 39. Koebel CM, Vermi W, Swann JB, Zerafa N, Rodig SJ, Old LJ, et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature 2007;450:903–7. 40. Boshoff C, Weiss R. AIDS-related malignancies. Nat Rev Cancer 2002;2:373–82. 41. Moon EJ, Jeong CH, Jeong JW, Kim KR, Yu DY, Murakami S, et al. Hepatitis B virus X protein induces angiogenesis by stabilizing hypoxia-inducible factor-1alpha. FASEB J 2004;18:382–4. 42. Botelho MC, Oliveira PA, Lopes C, Correia da Costa JM, Machado JC. Urothelial dysplasia and inflammation induced by Schistosoma Clinical Cancer Research Downloaded from clincancerres.aacrjournals.org on May 20, 2013. © 2013 American Association for Cancer Research. Published OnlineFirst March 27, 2013; DOI: 10.1158/1078-0432.CCR-12-3661 Infection and Cancer 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. haematobium total antigen instillation in mice normal urothelium. Urol Oncol 2011;29:809–14. Thomas JE, Bassett MT, Sigola LB, Taylor P. Relationship between bladder cancer incidence, Schistosoma haematobium infection, and geographical region in Zimbabwe. Trans R Soc Trop Med Hyg 1990; 84:551–3. Fassi FL, Mak TN, Laube B, Brinkmann V, Ogilvie LA, Mollenkopf H, et al. Prevalence of Propionibacterium acnes in diseased prostates and its inflammatory and transforming activity on prostate epithelial cells. Int J Med Microbiol 2011;301:69–78. Compare D, Nardone G. Contribution of gut microbiota to colonic and extracolonic cancer development. Dig Dis 2011;29:554–61. Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci U S A 1975;72:3666–70. Wiemann B, Starnes CO. Coley's toxins, tumor necrosis factor and cancer research: a historical perspective. Pharmacol Ther 1994;64: 529–64. Moore RJ, Owens DM, Stamp G, Arnott C, Burke F, East N, et al. Mice deficient in tumor necrosis factor-alpha are resistant to skin carcinogenesis. Nat Med 1999;5:828–31. Rigano R, Buttari B, Profumo E, Ortona E, Delunardo F, Margutti P, et al. Echinococcus granulosus antigen B impairs human dendritic cell differentiation and polarizes immature dendritic cell maturation towards a Th2 cell response. Infect Immun 2007;75:1667–78. Gomez-Garcia L, Lopez-Marin LM, Saavedra R, Reyes JL, RodriguezSosa M, Terrazas LI. Intact glycans from cestode antigens are involved in innate activation of myeloid suppressor cells. Parasite Immunol 2005;27:395–405. Uzonna JE, Kaushik RS, Gordon JR, Tabel H. Immunoregulation in experimental murine Trypanosoma congolense infection: anti-IL-10 antibodies reverse trypanosome-mediated suppression of lymphocyte proliferation in vitro and moderately prolong the lifespan of genetically susceptible BALB/c mice. Parasite Immunol 1998;20:293–302. Carrow EW, Domer JE. Immunoregulation in experimental murine candidiasis: specific suppression induced by Candida albicans cell wall glycoprotein. Infect Immun 1985;49:172–81. Wherry EJ. T cell exhaustion. Nat Immunol 2011;12:492–9. Coley WB. The treatment of malignant tumors by repeated inoculations of erysipelas. With a report of ten original cases. 1893. Clin Orthop Relat Res 1991;3–11. Alexandroff AB, Nicholson S, Patel PM, Jackson AM. Recent advances in bacillus Calmette-Guerin immunotherapy in bladder cancer. Immunotherapy 2010;2:551–60. Krone B, Kolmel KF, Henz BM, Grange JM. Protection against melanoma by vaccination with Bacille Calmette-Guerin (BCG) and/or vaccinia: an epidemiology-based hypothesis on the nature of a melanoma risk factor and its immunological control. Eur J Cancer 2005;41:104–17. Mastrangelo G, Krone B, Fadda E, Buja A, Grange JM, Rausa G, et al. Does yellow fever 17D vaccine protect against melanoma? Vaccine 2009;27:588–91. Oliveira EC, Leite MS, Miranda JA, Andrade AL, Garcia SB, Luquetti AO, et al. Chronic Trypanosoma cruzi infection associated with low incidence of 1,2-dimethylhydrazine-induced colon cancer in rats. Carcinogenesis 2001;22:737–40. Harnett W, Harnett MM. Helminth-derived immunomodulators: can understanding the worm produce the pill? Nat Rev Immunol 2010;10: 278–84. van den Biggelaar AH, van RR, Rodrigues LC, Lell B, Deelder AM, Kremsner PG, et al. Decreased atopy in children infected with Schistosoma haematobium: a role for parasite-induced interleukin-10. Lancet 2000;356:1723–7. O'Garra A, Barrat FJ, Castro AG, Vicari A, Hawrylowicz C. Strategies for use of IL-10 or its antagonists in human disease. Immunol Rev 2008;223:114–31. Ercolini AM, Miller SD. The role of infections in autoimmune disease. Clin Exp Immunol 2009;155:1–15. Butterfield LH, Ribas A, Dissette VB, Amarnani SN, Vu HT, Oseguera D, et al. Determinant spreading associated with clinical response in www.aacrjournals.org 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. dendritic cell-based immunotherapy for malignant melanoma. Clin Cancer Res 2003;9:998–1008. Ratliff TL, Ritchey JK, Yuan JJ, Andriole GL, Catalona WJ. T-cell subsets required for intravesical BCG immunotherapy for bladder cancer. J Urol 1993;150:1018–23. Mizzen L. Immune responses to stress proteins: applications to infectious disease and cancer. Biotherapy 1998;10:173–89. Springer GF. Immunoreactive T and Tn epitopes in cancer diagnosis, prognosis, and immunotherapy. J Mol Med (Berl) 1997;75: 594–602. Li Q, Anver MR, Butcher DO, Gildersleeve JC. Resolving conflicting data on expression of the Tn antigen and implications for clinical trials with cancer vaccines. Mol Cancer Ther 2009;8:971–9. Pfister M, Gottstein B, Cerny T, Cerny A. Immunodiagnosis of echinococcosis in cancer patients. Clin Microbiol Inf 1999;5:693–7. Hjelm F, Carlsson F, Getahun A, Heyman B. Antibody-mediated regulation of the immune response. Scand J Immunol 2006;64: 177–84. Cox FE. Concomitant infections, parasites and immune responses. Parasitology 2001;122 Suppl:S23–38. Fisher B, Saffer EA, Fisher ER. Comparison of concomitant and sinecomitant tumor immunity. Proc Soc Exp Biol Med 1970;135: 68–71. Honda K, Littman DR. The microbiome in infectious disease and inflammation. Annu Rev Immunol 2012;30:759–95. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012; 12:253–68. Tartour E, Pere H, Maillere B, Terme M, Merillon N, Taieb J, et al. Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev 2011;30:83–95. Bukowski JA, Lewis RJ. Is the hygiene hypothesis an example of hormesis?Nonlinearity Biol Toxicol Med 2003;1:155–66. Quigley EM. Prebiotics and probiotics; modifying and mining the microbiota. Pharmacol Res 2010;61:213–8. Tuohy KM, Probert HM, Smejkal CW, Gibson GR. Using probiotics and prebiotics to improve gut health. Drug Discov Today 2003;8: 692–700. Movahedi K, Guilliams M, Van den Bossche J, Van den Bergh R, Gysemans C, Beschin A, et al. Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cellsuppressive activity. Blood 2008;111:4233–44. Voisin MB, Buzoni-Gatel D, Bout D, Velge-Roussel F. Both expansion of regulatory GR1þ CD11bþ myeloid cells and anergy of T lymphocytes participate in hyporesponsiveness of the lung-associated immune system during acute toxoplasmosis. Infect Immun 2004;72: 5487–92. Hunter CA, Yu D, Gee M, Ngo CV, Sevignani C, Goldschmidt M, et al. Cutting edge: systemic inhibition of angiogenesis underlies resistance to tumors during acute toxoplasmosis. J Immunol 2001; 166:5878–81. Kaiser J. Will an aspirin a day keep cancer away? Science 2012;337: 1471–3. Scott AM, Wolchok JD, Old LJ. Antibody therapy of cancer. Nat Rev Cancer 2012;12:278–87. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012;12:265–77. Heimburg-Molinaro J, Lum M, Vijay G, Jain M, Almogren A, Rittenhouse-Olson K. Cancer vaccines and carbohydrate epitopes. Vaccine 2011;29:8802–26. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature 2011;480:480–9. Erdman SE, Rao VP, Olipitz W, Taylor CL, Jackson EA, Levkovich T, et al. Unifying roles for regulatory T cells and inflammation in cancer. Int J Cancer 2010;126:1651–65. Patyar S, Joshi R, Byrav DS, Prakash A, Medhi B, Das BK. Bacteria in cancer therapy: a novel experimental strategy. J Biomed Sci 2010; 17:21. Clin Cancer Res; 19(11) June 1, 2013 Downloaded from clincancerres.aacrjournals.org on May 20, 2013. © 2013 American Association for Cancer Research. OF7 Published OnlineFirst March 27, 2013; DOI: 10.1158/1078-0432.CCR-12-3661 Oikonomopoulou et al. 88. Liu WM, Fowler DW, Gravett AM, Smith P, Dalgleish AG. Supernatants from lymphocytes stimulated with Bacillus Calmette-Guerin can modify the antigenicity of tumours and stimulate allogeneic T-cell responses. Br J Cancer 2011;105:687–93. 89. Ahirwar DK, Manchanda PK, Mittal RD, Bid HK. BCG response prediction with cytokine gene variants and bladder cancer: where we are? J Cancer Res Clin Oncol 2011;137:1729–38. 90. Springer GF, Desai PR, Spencer BD, Tegtmeyer H, Carlstedt SC, Scanlon EF. T/Tn antigen vaccine is effective and safe in preventing recurrence of advanced breast carcinoma. Cancer Detect Prev 1995; 19:374–80. 91. Jouvin MH, Kinet JP. Trichuris suis ova: testing a helminth-based therapy as an extension of the hygiene hypothesis. J Allergy Clin Immunol 2012;130:3–10. 92. Summers RW, Elliott DE, Urban JF Jr, Thompson RA, Weinstock JV. Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology 2005;128:825–32. OF8 Clin Cancer Res; 19(11) June 1, 2013 93. Fox JG, Beck P, Dangler CA, Whary MT, Wang TC, Shi HN, et al. Concurrent enteric helminth infection modulates inflammation and gastric immune responses and reduces helicobacter-induced gastric atrophy. Nat Med 2000;6:536–42. 94. Akgul H, Tez M, Unal AE, Keskek M, Sayek I, Ozcelik T. Echinococcus against cancer: why not? Cancer 2003;98:1999–2000. 95. Hibbs JB Jr, Lambert LH Jr, Remington JS. Resistance to murine tumors conferred by chronic infection with intracellular protozoa, Toxoplasma gondii and Besnoitia jellisoni. J Infect Dis 1971;124: 587–92. 96. Lemmer LB, Fripp PJ. Schistosomiasis and malignancy. S Afr Med J 1994;84:211–5. 97. Fried B, Reddy A, Mayer D. Helminths in human carcinogenesis. Cancer Lett 2011;305:239–49. 98. Porta C, Riboldi E, Sica A. Mechanisms linking pathogensassociated inflammation and cancer. Cancer Lett 2011;305: 250–62. Clinical Cancer Research Downloaded from clincancerres.aacrjournals.org on May 20, 2013. © 2013 American Association for Cancer Research.