<<

Journal of Bacteriology and Virology 2015. Vol. 45, No. 3 p.179 – 188 http://dx.doi.org/10.4167/jbv.2015.45.3.179 Review Article

Influence of the Host Factors on Human Papillomavirus and Efficacy

* Sung-Hwa Sohn and Jae-Hwan Nam

Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Korea

Human papillomavirus (HPV) is associated with cervical cell changes, genital warts, recurrent respiratory papillomatosis, laryngeal papillomatosis, head and neck , and cervical cancer. Two commercial HPV have successfully been made available in the clinical field. This review covers the progress of cervical disease by understanding the nature of HPV infection, as well as the relationship between the host factors and HPV vaccine effectiveness. Among these host factors, microbiota has been revealed to influence the development and function of both the innate and adaptive immune systems. Therefore, the composition of the microbiome may ultimately affect vaccine efficacy. Understating the relationship between host factors and HPV infection/vaccine efficacy may prove to be useful in earlier diagnosis, as well as disease prophylaxis. Key Words: HPV, Vaccine efficacy, Probiotics, Microbiota

sequent development of cervical have been exten- INTRODUCTION sively studied, there are many unknowns in the relationship between the host factors (e.g. genetic polymorphism, micro- Humans are constantly exposed to pathogenic micro- biome, disease status, and immune system) and HPV vaccine organisms, including viruses, bacteria, and fungi. Among effectiveness. Among the various host factors affecting these , human papillomavirus (HPV) is the vaccine efficacy, the role of the microbiome has now been most common sexually transmitted infection worldwide (1). established (4, 5). In this review, we will summarize the Currently, two HPV commercial vaccines have been recent data on the relationship between the host factors, approved for use in the clinical field. These are termed especially microbiota composition, and both HPV vaccine '' and '', which cover 4 (6, 11, 16 and 18) efficacy and HPV infection. and 2 (16 and 18) HPV types, respectively (2). In December 2014, Gardasil 9 was approved in the USA, which covers 9 Basics of HPV infection HPV types (6, 11, 16, 18, 31, 33, 45, 52, and 58) (3). The characters of these HPV vaccines are summarized in Table HPV is a circular double-stranded DNA (dsDNA) virus 1. While the mechanism of HPV infection and the sub- for which more than 200 different types have been identified

Received: May 30, 2015/ Revised: June 10, 2015/ Accepted: June 25, 2015 *Corresponding author: Jae-Hwan Nam, Ph.D. Department of Biotechnology, The Catholic University of Korea, 43-1 Yeokgok 2-dong, Wonmi-gu, Bucheon, Gyeonggi-do, 420-743, Korea. Phone: +82-2-2164-4852, Fax: +82-2-2164-4917, e-mail: [email protected] **This study was supported by a grant from the Korean Healthcare Technology R&D project of the Ministry of Health & Welfare (A103001 and HI13C0826), and by a grant from Gyunggi Regional Research Center (GRRC) of the Catholic University of Korea.

○CC This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/license/by-nc/3.0/).

179

180 S-H Sohn and J-H Nam

Table 1. Characteristics of anti-HPV vaccines

Bivalent vaccine Quadrivalent vaccine Ninevalent vaccine Commercial name Cervarix® Gardasil® Gardasil® 9 HPV 6/ 11/ 16/ 18/ 31/ 33/ HPV types HPV 16/ 18 HPV 6/ 11/ 16/ 18 45/ 52/ 58 Trichoplusia ni (Hi 5) insect Saccharomyces cerevisiae Saccharomyces cerevisiae Producer cells cell line infected with L1 expressing L1 expressing L1 recombinant baculovirus Girls and women: 9~26 years: Administration Girls and women: 9~14 years: Girls and women: 9~13 years: 0, 2, 6 months schedule 0, 1, 6 months 0, 2, 6 months Boys: 9~15 years: 0, 2, 6 months Aluminum hydroxide and Aluminum hydroxyphosphate Aluminum hydroxyphosphate Adjuvant 3-O-deacylated-4-monophosphoryl sulfate sulfate or AAHS lipid A

and categorized into high- and low risk (6). HPV has a non- (L1 and L2) and three oncoproteins (E5, E6 and E7) (17, enveloped, icosahedral capsid, and infects only mucosal or 18). The major capsid L1 self-assembles into virus- cutaneous epithelium (7). The fifteen types of HPV classified like particles (VLPs), which are in tertiary or native form as high risk include HPV16, 18, 31, 33, 35, 39, 45, 51, 52, and are assembled as multimers to generate neutralizing 56, 58, 59, 68, 73, and 82 (8). In particular, HPV 16 and 18 antibodies. HPV L1 VLP vaccines have been developed are known to cause up to 70% of cervical cancers (9). The commercially (Table 1). VLP vaccines are highly immuno- low risk HPV types include types 6, 11, 42, 43, and 44, genic and induce HPV neutralizing antibodies, which persist which are involved in the development of cervical cell for at least 5 years after . The HPV E5 protein changes, genital warts, recurrent respiratory papillomatosis, suppresses the expression of MHC class I and antigen and laryngeal papillomatosis (10~12). processing via the transporter associated with the antigen Cervical cancer, the second most prevalent cancer among processing pathway (19, 20). HPV E6 ubiquitinates (ubi- women, is associated with HPV infection (13). To detect quitin mediated degradation) the p53 tumor suppressor potentially dangerous HPV strains and to defend against protein (21), whereas E7 ubiquitinates the retinoblastoma infection, humans have developed effective innate and (pRb) tumor suppressor protein (22), indicating that these adaptive immune systems that minimize the impact of two may have oncogenic effects. In addition, the pathogens on human health. On the other hand, HPV uses HPV E6 protein causes downregulation of the expression specific life strategies to enable its survival and persistence of interferon-related and transcription factor NF-κB within the epithelial cells, including diverse mechanisms to (23). NF-κB activates a local immune response (e.g., MHC evade host immune surveillance via programming for natural class I, interleukins and GM-CSF) and an IFN-β promoter cell death, which causes the progeny virions only to be (24). Inhibition of NF-κB may help the HPV virus to escape released in high quantities from terminally differentiated immune recognition. The HPV E7 protein blocks cell cycle epithelial cells (14). HPV infects only mucosal or cutaneous regulatory genes are required for G1/S transition and DNA epithelium. In addition, HPV induces a slow and weak replication. These proteins (E5, E6 and E7) are also able to immune response, which inhibits interferon synthesis (15, break antigen presentation (25) (Fig. 1). Therefore, these 16). three proteins also play roles in the evasion of the innate The HPV genome encodes two structural capsid proteins and adaptive immune system, which creates a favorable

Host Factor Ramifications on 181

Figure 1. A schematic diagram of a possible mode of the oncogenic action of human papillomavirus (HPV). HPV infection into a host tissue leads to oncogenic processes. HPV E5 protein suppresses the expression of MHC class I and antigen processing. HPV E6 protein binds p53 tumor suppressor protein and this binding blocks p53 defense mechanisms, as well as p53-induced antiviral activity (down-regulates the expression of the antiviral interferon-related genes and the transcription factor NF-κB), through cell cycle arrest and apoptosis of the infected cell by degradation of p53 via ubiquitination. HPV E7 protein binds pRb cell cycle regulatory protein, which can block cell cycle arrest by degradation of Rb via ubiquitination. Therefore, these proteins play roles in tumor promotion. These HPV E5, E6, and E7 proteins are able to impair the innate and acquired immune response, which play roles in the evasion of the host immune system. MHC class I, major histcompatibility complex class I; pRb, retinoblastoma protein; Ag, antigen; IFN, interferon gamma; NF-κB, nuclear factor-κB.

environment for viral replication (26). It is thought that E6 1. Impact of host genetic factors on HPV infection and E7, from the low risk HPV types, may have weaker binding to their target proteins compared to the high risk Population based association studies (genetic polymor- types (27). phisms) often estimate the risk of developing a certain disease in carriers and noncarriers of a particular genetic Factors affecting HPV infection and vaccine polymorphism (30). The population genetic background contributes to the susceptibility to developing cervical cancer Many factors affect HPV infection, including sex, estrogen after HPV infection. The p53 Pro/p53 Arg polymorphism hormones, microbiome, genetics, epigenetic phenomena, age, should influence the outcome of HPV infection, because nutrition, immune system, and infectious and autoimmune the presence of heterozygous p53 Pro is more resistant than diseases (28, 29). p53 Arg to E6-mediated degradation. Those with the p53 Arg allele, in the absence of the p53 Pro allele, are there- fore at increased risk of developing cervical cancer (31).

182 S-H Sohn and J-H Nam

Moreover, polymorphism in the proinflammatory cytokine 3. Impact of host immune factor on intestinal micro- Tumor necrosis factor-α (TNF-α) also causes increased biota the risk of HPV associated cervical cancer in the presence of HPV. TNF-α-308 G/A polymorphism may play a role in Recognition of microorganisms by the innate immune TNF-α production and the inflammatory response during system involves pattern recognition receptors (PRRs), such the course of cervical cancer. In particular, the A allele is as nucleotide binding oligomerization domain (Nod) like associated with an increased risk of invasive cervical cancer receptors (NLRs) and Toll like receptors (TLRs), which dis- (32, 33). Individuals with the TNF-α-308 G/A and -238 criminate pathogen associated molecular patterns (PAMPs) G/A promoter genotypes are at increased risk of developing or danger associated molecular patterns (DAMPs) (52~54). inflammatory, infectious and (33). Extracellular contaminants are recognized by TLRs, whereas These results suggest that host genetic polymorphisms can surveillance of the cytoplasm is the work of NLRs, recog- influence the nature and extent of HPV-mediated disease. nized by their Nods (52, 55). Among the various TLRs, TLR4 recognizes pathogen bacterial LPS, but is not stimu- 2. Impact of host microbiome on metabolic disorder lated by commensal microflora, which have detoxified forms and chronic inflammatory disease of LPS, dephosphorylated by an intestine-specific isoform The interplay between HPV, the gut epithelium, and host of alkaline phosphatase (36). TLR5 recognizes bacterial innate defense responses is among the most critical factors flagella, and its signaling can stimulate CD11bhighCD11chigh determining the fate of HPV and disease out- DCs to express Raldh2, which catalyzes the conversion of comes. Recently, gut microbiota were found to contribute vitamin A to retinoic acid. Retinoic acid can then cause B to many host physiological processes and disease (34, 35). cells to differentiate into IgA producing plasma cells (56). Theses intestinal microbiota make important contributions Secreted IgA is required for keeping commensal bacteria to the health of their hosts by protect against pathogens via that can determine the nature of bacteria. IgA regulates the appropriate development of the immune system and syn- ecological balance of microbiota, which regulates the com- thesis of essential vitamins (35, 36). It should also be noted position and character of microbiota. IgA protects against that these essential vitamins, such as vitamin A and D, are viruses from the epithelium by binding, rather than cellular also important for immune function and vaccine effectiveness neutralization. Furthermore, IgA, an adaptive response effector, (37). In addition, there is an abundance of NK, Th17 and is crucial for the compartmentalization of the microbiota to Treg cells in the intestinal mucosa, and their accumulation the intestinal lumen (57) (Fig. 2). However, Tlr5-/- mice and function are affected by the presence of the microbiota exhibit similar changes in the microbiota shape as ob/ob (38). Infant gut microbiota has a relatively simple com- mice (leptin gene deficiency). The Tlr5-/- phenotype is due position, which is affected in large part by the maternal to changes in the microbiota, causing hyperphagia and microbiota due to the delivery method, such as cesarean or metabolic syndromes, including insulin resistance, hyper- vaginal birth (39). Moreover, community and function of lipidemia, and obesity (43). TLR9 recognizes the viral or the microbiota can change owing to numerous variables, bacterial dsDNA derived CpG motifs, but its transcription including lifestyle, diet, hygiene, and use of antibiotics (40). is inhibited by recombinant HPV 16 E6 and E7 (26). Two of the main types of intestinal microbes, the Fir- TLR2 and Nod2 are both required for the transcriptional micutes and Bacteroidetes, have also been linked to obesity induction of pro IL-1β and the priming of the , and metabolic disorders (41~43). Furthermore, microbiota which is mediated by the upregulation of NLRP3 (58). composition is also related to incidence of chronic inflam- Inflammasome activation is thought to require two signals, matory diseases, such as allergic conditions, chronic colitis, the first involving the sensing of conserved PAMPs which and autoimmune disorders (44~51). induce expression of certain inflammasome components and

Host Factor Ramifications on Vaccination 183

Figure 2. A schematic diagram of possible mode for the relationship between the microbiota and the immune system in the intestinal lamina propria. The microbiota is composed of a variety of bacteria with different characters. The microbiota influence the development and function of both the innate and adaptive immune systems. The intestinal microflora has TLR5 ligans such as flagellin. TLR5 signaling stimulates CD11bhighCD11chigh DCs, which can catalyze the conversion of vitamin A to retinoic acid; this causes B cells to differentiate into IgA producing plasma cells. Secreted IgA regulates the ecological balance of microbiota, which regulates the composition and character of microbiota. Ultimately, IgA induces compartmentalization of the microbiota to the intestinal lumen, and it contributes to protection from viral infections. HPV, human papillomavirus; TLR5, toll like receptor 5; DC, dendritic cell; IgA, immunoglobulin A; Raldh2, retinaldehyde dehydrogenase 2.

cytokines, and the second involving disruption of cellular bacteria that have proinflammatory activities (46, 62, 63). membranes or signaling pathways (59). Cytosolic dsDNA Nod2-/- mice are susceptible to pathogen infection because from intracellular pathogens such as HPV is sensed by of an intrinsic defect in their ability to kill the bacteria. In AIM2, which triggers formation of the inflammasome com- addition, these mice also have increased populations of plex with ASC to activate 1-mediated processing commensal bacteria in the intestine, suggesting that NOD2 of IL-1β and IL-18 (60, 61). Recent studies have suggested plays a crucial role in regulating between that inflammasome deficiency may be associated with bacterial flora and innate immunity (64). Moreover, NLRP6 stimulator-induced inflammation, inducing tumorigenesis deficient mice exhibited increased members of Prevotell- in the intestine and chemical colitis, due to alterations in aceae and TM7, and reduced members of Lactobacillus, the microbiota and the emergence of normally suppressed which caused increased intestinal inflammatory cell recruit-

184 S-H Sohn and J-H Nam ment and susceptibility to chemically induced colitis. These malnutrition, or hygiene (66), which may easily affect the mice phenotypes resembled ASC-/- mice (46). Therefore, microbiota composition. Consequently, based on these well- host immune factors may drive disease-specific alterations in founded conjectures, microbiota may affect vaccine responses the microbiota, which in turn may promote the development indirectly or directly through action on various immune of disease in the host. responses, such as development of CD4+ T cells (67). Immunization is necessary to prevent morbidity and mor- 4. Impact of host microbiome on HPV vaccine efficacy tality from infectious diseases, such as HPV. Actually, the The human intestinal tract harbors a large number of main goal of vaccination is to elicit memory B cell responses species which intimately interact with nutrition status, gen- and CD4+ T cell-mediated neutralizing immune effector etics, metabolism, and the immune system (65). The micro- responses. B cell memory response to antigen recall is an biota ultimately contribute to host physiology via fostering important factor for the long-term efficacy of vaccine- normal development of the immune system and influencing induced humoral protection. The protection achieved by the ensuing immune responses. Interestingly, vaccines often HPV vaccines against incident HPV infection and HPV- have lower effectiveness than expected when used in associated pathology is mediated via serum neutralizing developing countries, possibly due to intestinal dysbiosis, IgG (68). By the way, recent studies have shown a trend

Table 2. Summary of the impact of probiotics on vaccine efficacy

Probiotics Vaccines Biological effects Lacrobacillus casei D x RRV rhesus-human LGG has an immunostimulatinq effect on rotavirus vaccination strain GG (LGG) reassortant via enhance IgA production and release of interferon γ. LGG stimulated IgA sASC responses against S. typhi Ty21a. LGG or Lactococcus lactis Salmonella typhi L. lactis has increase CR3 receptor expression on the neutrophils (L. lactis) Ty21a vaccine which can influence the non-specific immune response. LGG and Bifidobacterium lactis LGG and BB12 acted as an immunostimulant for AttHRV AttHRV vaccine BB12 (BB12) vaccine via modulated DC activation and responses. L. lactis, Lactobacillus plantarum These bacteria have induction of Th1 response in eczema and Allergy vaccines or Bifidobacterium bifidum asthma models. Mycobacterium vaccae Heat-killed M. vaccae induces Treg cells and decreases airway Allergy vaccines (M. vaccae) inflammation in or asthma murine model. Bifidobacterium breve strain A trend towards increased serum IgG after administration of Yakult (BBG-01) BBG-01 Bifidobacterium longum BL999 BL999 and LPR were improved HepB surface antibody (BL999) and Lactobacillus B vaccine responses in subjects. rhamnosus LPR (LPR) Lactobacillus paracasei ssp. Diphtheria, tetanus and LF19 was increased the capacity to raise immune responses in F19 (LF19) Hib vaccines infants breastfed < 6 months. Live attenuated influenza LGG LGG was increased protective hemagglutinin inhibition titers. vaccines L. fermentum A trend towards increased TNF-α, total IgG and IgM, and Inactivated CECT5716 (LCECT5716) influenza-specific IgA after administration of LCECT5716. BB12 and L. paracasei BB12 and L431 were elevated influenza specific IgG levels and Inactivated influenza vaccine 431 (L431) influenza specific IgA responses.

Host Factor Ramifications on Vaccination 185 towards increased serum IgG after administration of pro- Ghissassi F, et al. Carcinogenicity of human papilloma- biotics (69, 70). However, probiotic formulations may not viruses. Lancet Oncol 2005;6:204. improve the response to a particular vaccine (71). The recent 2) Stanley M, Lowy DR, Frazer I. Chapter 12: Prophylactic data regarding the relationship between probiotic treatment HPV vaccines: underlying mechanisms. Vaccine 2006; and vaccine efficacy are summarized in Table 2. Taken 24. together, these data indicate that the effects of probiotics on 3) US Food and Drug Administaration. FDA approves Gar- dasil 9 for prevention of certain cancers caused by five vaccine efficacy may be dependent on the types of vaccines additional types of HPV, 2014. Available from: URL: (69, 70, 72~77). However, the detailed mechanisms urgently http://www.fda.gov/newsevents/newsroom/pressannou- need to be revealed. ncements/ucm426485.htm. Meanwhile, healthy vaginal microbiota is composed pri- 4) Valdez Y, Brown EM, Finlay BB. Influence of the marily of lactic acid producing bacteria, which contribute microbiota on vaccine effectiveness. Trends Immunol to women's health by maintaining a low pH in the vagina 2014;35:526-37. (5). Abnormal vaginal microbiota might act as a cofactor 5) Lee JE, Lee S, Lee H, Song YM, Lee K, Han MJ, et al. for the acquisition of HPV. Especially, L. gasseri and Association of the vaginal microbiota with human Gardnerella vaginalis were detected more frequently, along papillomavirus infection in a Korean twin cohort. PloS with a significantly higher diversity of the microbiota in One 2013;8:e63514. HPV-infected women than in HPV negative women (78). 6) zur Hausen H. Papillomaviruses in human cancers. Proc Ultimately, the composition of the vaginal microbiota may Assoc Am Physicians 1999;111:581-7. affect HPV infection and disease progression. 7) Dunne EF, Datta SD, E Markowitz L. A review of pro- phylactic human papillomavirus vaccines: recommen- CONCLUDING REMARKS dations and monitoring in the US. Cancer 2008;113: 2995-3003. As mentioned above, HPV infection may be at least 8) Muñoz N, Bosch FX, de Sanjosé S, Herrero R, partially dependent on vaginal microbiota composition, Castellsagué X, Shah KV, et al. Epidemiologic classifi- whereas the efficacy of the HPV vaccine may be dependent cation of human papillomavirus types associated with on the intestinal microbiota composition. However, to the cervical cancer. N Engl J Med 2003;348:518-27. best of our knowledge, direct evidence for the relationship 9) Burd EM. Human papillomavirus and cervical cancer. Clin Microbiol Rev 2003;16:1-17. between HPV vaccine efficacy and microbiota composition 10) Gissmann L, zur Hausen H. Partial characterization of has not yet been provided. Therefore, research of the viral DNA from human genital warts (Condylomata interaction between the host factors, especially microbiota, acuminata). Int J Cancer 1980;25:605-9. and HPV vaccine effectiveness is needed to enhance our 11) Lacey CJ, Lowndes CM, Shah KV. Chapter 4: Burden understanding of the HPV-related disease, cervical cancer, and management of non-cancerous HPV-related con- and to guarantee its prevention. ditions: HPV-6/11 disease. Vaccine 2006;24 Suppl 3:

S3/35-41. Footnotes 12) Gissmann L, Diehl V, Schultz-Coulon HJ, zur Hausen Disclosure: The authors have no conflicts of interest and H. Molecular cloning and characterization of human nothing to disclose. papilloma virus DNA derived from a laryngeal papil- loma. J Virol 1982;44:393-400. REFERENCES 13) Bray F, Sankila R, Ferlay J, Parkin DM. Estimates of cancer incidence and mortality in Europe in 1995. Eur 1) Cogliano V, Baan R, Straif K, Grosse Y, Secretan B, El J Cancer 2002;38:99-166.

186 S-H Sohn and J-H Nam

14) Stanley M. Immune responses to human papillomavirus. 29) Veldhuijzen NJ, Snijders PJ, Reiss P, Meijer CJ, van de Vaccine 2006;24 Suppl 1:S16-22. Wijgert JH. Factors affecting transmission of mucosal 15) Frazer I. Correlating immunity with protection for HPV human papillomavirus. Lancet Infect Dis 2010;10:862 infection. Int J Infect Dis 2007;11 Suppl 2:S10-6. -74. 16) Doorbar J. The papillomavirus life cycle. J Clin Virol 30) Demaria S, Pikarsky E, Karin M, Coussens LM, Chen 2005;32 Suppl 1:S7-15. YC, El-Omar EM, et al. Cancer and inflammation: 17) zur Hausen H. Papillomaviruses causing cancer: evasion promise for biologic therapy. J Immunother 2010;33: from host-cell control in early events in carcinogenesis. 335-51. J Natl Cancer Inst 2000;92:690-8. 31) Storey A, Thomas M, Kalita A, Harwood C, Gardiol D, 18) Chung CH, Gillison ML. Human papillomavirus in head Mantovani F, et al. Role of a p53 polymorphism in the and neck cancer: its role in pathogenesis and clinical development of human papillomavirus-associated cancer. implications. Clin Cancer Res 2009;15:6758-62. Nature 1998;393:229-34. 19) Tindle RW. Immune evasion in human papillomavirus- 32) Kohaar I, Thakur N, Salhan S, Batra S, Singh V, Sharma associated cervical cancer. Nat Rev Cancer 2002;2:59 A, et al. TNFalpha-308G/A polymorphism as a risk -65. factor for HPV associated cervical cancer in Indian 20) O'Brien PM, Campo MS. Papillomaviruses: a correlation population. Cell Oncol 2007;29:249-56. between immune evasion and oncogenicity? Trends 33) Duarte I, Santos A, Sousa H, Catarino R, Pinto D, Matos Microbiol 2003;11:300-5. A, et al. G-308A TNF-alpha polymorphism is associated 21) Scheffner M, Werness BA, Huibregtse JM, Levine AJ, with an increased risk of invasive cervical cancer. Howley PM. The E6 oncoprotein encoded by human Biochem Biophys Res Commun 2005;334:588-92. papillomavirus types 16 and 18 promotes the degra- 34) Littman DR, Pamer EG. Role of the commensal micro- dation of p53. Cell 1990;63:1129-36. biota in normal and pathogenic host immune responses. 22) Huh K, Zhou X, Hayakawa H, Cho JY, Libermann TA, Cell Host Microbe 2011;10:311-23. Jin J, et al. Human papillomavirus type 16 E7 onc- 35) Honda K, Littman DR. The microbiome in infectious oprotein associates with the cullin 2 ubiquitin ligase disease and inflammation. Annu Rev Immunol 2012;30: complex, which contributes to degradation of the retino- 759-95. blastoma tumor suppressor. J Virol 2007;81:9737-47. 36) Vaishnava S, Hooper LV. Alkaline phosphatase: keeping 23) Mantovani F, Banks L. The human papillomavirus E6 the peace at the gut epithelial surface. Cell Host Microbe protein and its contribution to malignant progression. 2007;2:365-7. Oncogene 2001;20:7874-87. 37) Mora JR, Iwata M, von Andrian UH. Vitamin effects on 24) Thanos D, Maniatis T. Virus induction of human IFN the immune system: vitamins A and D take centre stage. beta requires the assembly of an Nat Rev Immunol 2008;8:685-98. enhanceosome. Cell 1995;83:1091-100. 38) Littman DR, Rudensky AY. Th17 and regulatory T cells 25) DiMaio D, Liao JB. Human papillomaviruses and in mediating and restraining inflammation. Cell 2010; cervical cancer. Adv Virus Res 2006;66:125-59. 140:845-58. 26) Hasan UA, Bates E, Takeshita F, Biliato A, Accardi R, 39) Dominguez-Bello MG, Costello EK, Contreras M, Bouvard V, et al. TLR9 expression and function is Magris M, Hidalgo G, Fierer N, et al. Delivery mode abolished by the cervical cancer-associated human shapes the acquisition and structure of the initial micro- papillomavirus type 16. J Immunol 2007;178:3186-97. biota across multiple body habitats in newborns. Proc 27) Klingelhutz AJ, Roman A. Cellular transformation by Natl Acad Sci U S A 2010;107:11971-5. human papillomaviruses: lessons learned by comparing 40) Spor A, Koren O, Ley R. Unravelling the effects of the high- and low-risk viruses. Virology 2012;424:77-98. environment and host genotype on the gut microbiome. 28) Markle JG, Fish EN. SeXX matters in immunity. Trends Nat Rev Microbiol 2011;9:279-90. Immunol 2014;35:97-104. 41) Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V,

Host Factor Ramifications on Vaccination 187

Mardis ER, Gordon JI. An obesity-associated gut micro- G, Kim YG, et al. Intracellular NOD-like receptors in biome with increased capacity for energy harvest. Nature innate immunity, infection and disease. Cell Microbiol 2006;444:1027-31. 2008;10:1-8. 42) Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, 54) Yoneyama M, Fujita T. Structural mechanism of RNA Duncan A, Ley RE, et al. A core gut microbiome in recognition by the RIG-I-like receptors. Immunity 2008; obese and lean twins. Nature 2009;457:480-4. 29:178-81. 43) Vijay-Kumar M, Aitken JD, Carvalho FA, Cullender TC, 55) Swanson MS, Molofsky AB. Autophagy and inflam- Mwangi S, Srinivasan S, et al. Metabolic syndrome and matory cell death, partners of innate immunity. Auto- altered gut microbiota in mice lacking Toll-like receptor phagy 2005;1:174-6. 5. Science 2010;328:228-31. 56) Uematsu S, Fujimoto K, Jang MH, Yang BG, Jung YJ, 44) Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Nishiyama M, et al. Regulation of humoral and cellular Umesaki Y, et al. Gut-residing segmented filamentous gut immunity by lamina propria dendritic cells ex- bacteria drive autoimmune arthritis via T helper 17 cells. pressing Toll-like receptor 5. Nat Immunol 2008;9:769 Immunity 2010;32:815-27. -76. 45) Garrett WS, Lord GM, Punit S, Lugo-Villarino G, 57) Macpherson AJ, Geuking MB, Slack E, Hapfelmeier S, Mazmanian SK, Ito S, et al. Communicable ulcerative McCoy KD. The habitat, double life, citizenship, and colitis induced by T-bet deficiency in the innate immune forgetfulness of IgA. Immunol Rev 2012;245:132-46. system. Cell 2007;131:33-45. 58) Kim DJ, Park JH, Franchi L, Backert S, Núñez G. The 46) Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss Cag pathogenicity island and interaction between TLR2 CA, Booth CJ, et al. NLRP6 inflammasome regulates /NOD2 and NLRP3 regulate IL-1beta production in colonic microbial ecology and risk for colitis. Cell 2011; Helicobacter pylori infected dendritic cells. Eur J 145:745-57. Immunol 2013;43:2650-8. 47) Scher JU, Abramson SB. The microbiome and rheu- 59) Mariathasan S, Monack DM. Inflammasome adaptors matoid arthritis. Nat Rev Rheumatol 2011;7:569-78. and sensors: intracellular regulators of infection and 48) Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. inflammation. Nat Rev Immunol 2007;7:31-40. Proinflammatory T-cell responses to gut microbiota 60) Dombrowski Y, Peric M, Koglin S, Kammerbauer C, promote experimental autoimmune encephalomyelitis. Göss C, Anz D, et al. Cytosolic DNA triggers inflam- Proc Natl Acad Sci U S A 2011;108 Suppl 1:4615-22. masome activation in keratinocytes in psoriatic lesions. 49) Berer K, Mues M, Koutrolos M, Rasbi ZA, Boziki M, Sci Transl Med 2011;3:82ra38. Johner C, et al. Commensal microbiota and myelin 61) Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind autoantigen cooperate to trigger autoimmune demyeli- F, Horvath G, Caffrey DR, et al. AIM2 recognizes nation. Nature 2011;479:538-41. cytosolic dsDNA and forms a caspase-1-activating in- 50) Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan flammasome with ASC. Nature 2009;458:514-8. L, Stonebraker AC, et al. Innate immunity and intes- 62) Chen GY, Liu M, Wang F, Bertin J, Núñez G. A tinal microbiota in the development of Type 1 diabetes. functional role for Nlrp6 in intestinal inflammation and Nature 2008;455:1109-13. tumorigenesis. J Immunol 2011;186:7187-94. 51) Kriegel MA, Sefik E, Hill JA, Wu HJ, Benoist C, Mathis 63) Allen IC, TeKippe EM, Woodford RM, Uronis JM, D. Naturally transmitted segmented filamentous bacteria Holl EK, Rogers AB, et al. The NLRP3 inflammasome segregate with diabetes protection in nonobese diabetic functions as a negative regulator of tumorigenesis during mice. Proc Natl Acad Sci U S A 2011;108:11548-53. colitis-associated cancer. J Exp Med 2010;207:1045-56. 52) Iwasaki A, Medzhitov R. Toll-like receptor control of 64) Petnicki-Ocwieja T, Hrncir T, Liu YJ, Biswas A, the adaptive immune responses. Nat Immunol 2004;5: Hudcovic T, Tlaskalova-Hogenova H, et al. Nod2 is 987-95. required for the regulation of commensal microbiota in 53) Franchi L, Park JH, Shaw MH, Marina-Garcia N, Chen the intestine. Proc Natl Acad Sci U S A 2009;106:

188 S-H Sohn and J-H Nam

15813-8. double-blind trial. Pediatr Allergy Immunol 2006;17: 65) Hooper LV, Littman DR, Macpherson AJ. Interactions 416-21. between the microbiota and the immune system. Science 72) Isolauri E, Joensuu J, Suomalainen H, Luomala M, 2012;336:1268-73. Vesikari T. Improved immunogenicity of oral D x RRV 66) Qadri F, Bhuiyan TR, Sack DA, Svennerholm AM. reassortant rotavirus vaccine by Lactobacillus casei Immune responses and protection in children in devel- GG. Vaccine 1995;13:310-2. oping countries induced by oral vaccines. Vaccine 2013; 73) Fang H, Elina T, Heikki A, Seppo S. Modulation of 31:452-60. humoral immune response through probiotic intake. 67) Huda MN, Lewis Z, Kalanetra KM, Rashid M, Ahmad FEMS Immunol Med Microbiol 2000;29:47-52. SM, Raqib R, et al. Stool microbiota and vaccine 74) Vlasova AN, Chattha KS, Kandasamy S, Liu Z, Esseili responses of infants. Pediatrics 2014;134:e362-72. M, Shao L, et al. Lactobacilli and bifidobacteria promote 68) Bryan JT, Jansen KU, Lowe RS, Fife KH, McClowry T, immune homeostasis by modulating innate immune Glass D, et al. Human papillomavirus type 11 neutra- responses to human rotavirus in neonatal gnotobiotic lization in the athymic mouse xenograft system: cor- pigs. PloS One 2013;8:e76962. relation with virus-like particle IgG concentration. J 75) Moingeon P. Adjuvants for allergy vaccines. Hum Vaccin Med Virol 1997;53:185-8. Immunother 2012;8:1492-8. 69) Matsuda F, Chowdhury MI, Saha A, Asahara T, Nomoto 76) Zuany-Amorim C, Manlius C, Trifilieff A, Brunet LR, K, Tarique AA, et al. Evaluation of a probiotics, Bifido- Rook G, Bowen G, et al. Long-term protective and bacterium breve BBG-01, for enhancement of immuno- antigen-specific effect of heat-killed Mycobacterium genicity of an oral inactivated cholera vaccine and vaccae in a murine model of allergic pulmonary in- safety: a randomized, double-blind, placebo-controlled flammation. J Immunol 2002;169:1492-9. trial in Bangladeshi children under 5 years of age. 77) West CE, Gothefors L, Granström M, Käyhty H, Vaccine 2011;29:1855-8. Hammarström ML, Hernell O. Effects of feeding pro- 70) Soh SE, Ong DQ, Gerez I, Zhang X, Chollate P, Shek biotics during weaning on infections and antibody LP, et al. Effect of probiotic supplementation in the responses to diphtheria, tetanus and Hib vaccines. Pediatr first 6 months of life on specific antibody responses to Allergy Immunol 2008;19:53-60. infant Hepatitis B vaccination. Vaccine 2010;28:2577-9. 78) Gao W, Weng J, Gao Y, Chen X. Comparison of the 71) Kukkonen K, Nieminen T, Poussa T, Savilahti E, vaginal microbiota diversity of women with and without Kuitunen M. Effect of probiotics on vaccine antibody human papillomavirus infection: a cross-sectional study. responses in infancy--a randomized placebo-controlled BMC Infect Dis 2013;13:271.