Abstract

Considerable global cancer burden is attributable to infectious agents, or oncomicrobes, which provides an opportunity to limit cancer by vaccination. Hepatitis B virus and human papillomavirus vaccines exemplify this strategy, by significantly reducing cases of liver and cervical cancer, respectively, and highlight the potential of extending this approach to other oncomicrobial targets. However, to date, effective vaccines against other well-established oncomicrobes have not been developed, and novel oncomicrobes continue to emerge. In this review, we provide an overview of vaccination and vaccine design, with an emphasis on the key factors (vaccine type, antigen selection, administration route, and vaccination timing) to consider when developing oncomicrobial vaccines, and we summarize the current state and future directions of vaccination for several established and emerging oncomicrobes.

Vaccination is a cornerstone of public health, estimated to save 4 to 6 million lives globally per year.1 However, until recently, vaccination has mainly been directed toward preventing infectious disease. As we develop a greater appreciation of the complexity of host-microbe interactions, opportunities to apply vaccination to other aspects of public health, including cancer, have become evident.

In 1911, Peyton Rous provided the first evidence of infectious etiology of cancer by showing cell-free tumor filtrates could induce tumors by transmitting a virus.2,3 Since then, the ability of some microbes to exert oncogenic effects has become well established, with an estimated 13% to 18% of the global cancer burden attributable to known oncogenic infectious agents, or oncomicrobes.4 Hepatitis B virus (HBV), hepatitis C virus, human papillomavirus (HPV), Epstein-Barr virus (EBV), and the paradigmatic oncobacterium, Helicobacter pylori, are responsible for ∼97% of this estimated cancer burden attributable to infection,4,5 and the strong evidence of their oncogenicity has led to each being considered a class 1 carcinogen by the International Agency for Research on Cancer.6 These established agents and additional, recently identified oncomicrobes provide opportune vaccine targets for reduction of cancer burden. In this review, we summarize key concepts and principles relevant to oncomicrobe vaccine design, and highlight important successes of oncomicrobe vaccination, remaining challenges, and novel emerging targets.

Vaccination is exposure to an agent, or parts thereof, to evoke immunity and establish immune memory. This protects the vaccinated individual from future infection (through establishing circulating antibodies and effector cells, and/or by enabling a more rapid and effective anamnestic response) and, if the number of individuals vaccinated is sufficient to disrupt chains of transmission, also protects unvaccinated members of the population (ie, herd effect).7,8 To develop effective vaccines, key variables including vaccine type, antigen selection, administration route, and vaccine timing must be appropriately tailored for the specific microbe target of interest.

Vaccine type

Most clinically approved vaccines can be categorized into 5 types: live attenuated, killed/inactivated, recombinant protein, viral vector, and nucleic acid (Figure 1). Use of a whole organism as the immunogen is represented by the first 2 categories: live attenuated (“weakened”) vaccines and inactivated vaccines. Some live attenuated vaccines use microbes that are related to, but less virulent than, the pathogen of interest to induce crossprotective immunity. Indeed, such crossprotective application was used in the first application of vaccination, wherein cowpox vaccination was used to confer protection from smallpox9 (Box 1). Alternatively, the pathogen of interest can be directly attenuated, such as by desiccation or mutation during serial passaging in an atypical host.10,11 In some instances, live attenuated vaccines have natural and directed attenuation, such as Bacillus Calmette–Guérin (BCG) vaccines against tuberculosis (caused by Mycobacterium tuberculosis) that contain mutated Mycobacterium bovis virus with reduced invasion of lung tissue.12 

Figure 1.

The primary types of vaccines. Vaccine approaches can be categorized into either whole-cell (left) or subunit (right). Within approaches to whole-cell vaccination, the microbe of interest is attenuated (either by using a related, less virulent microbe that induces crossprotective immunity, or by directed attenuation of the microbe of interest), or killed/inactivated. Alternatively, vaccination can target a specific molecule of the microbe of interest in the case of subunit vaccination. Subunit vaccination can be performed through a variety of techniques, such as by use of recombinant protein or antigen-encoding mRNA LNP. Other vaccine modalities, such as toxoid vaccines (largely similar to native purified [with subsequent inactivation] or monomeric protein [with subsequent inactivation, or genetically inactivated]), protein-conjugate vaccines, and bacterial-vector vaccines are not shown. Figure was created in biorender.com. Holt Lab (2025). https://biorender.com/a6ynx7k

Figure 1.

The primary types of vaccines. Vaccine approaches can be categorized into either whole-cell (left) or subunit (right). Within approaches to whole-cell vaccination, the microbe of interest is attenuated (either by using a related, less virulent microbe that induces crossprotective immunity, or by directed attenuation of the microbe of interest), or killed/inactivated. Alternatively, vaccination can target a specific molecule of the microbe of interest in the case of subunit vaccination. Subunit vaccination can be performed through a variety of techniques, such as by use of recombinant protein or antigen-encoding mRNA LNP. Other vaccine modalities, such as toxoid vaccines (largely similar to native purified [with subsequent inactivation] or monomeric protein [with subsequent inactivation, or genetically inactivated]), protein-conjugate vaccines, and bacterial-vector vaccines are not shown. Figure was created in biorender.com. Holt Lab (2025). https://biorender.com/a6ynx7k

Close modal

Inactivated vaccines, or killed vaccines, use a whole organism rendered noninfectious (typically by heat or chemical treatment),11 making them considerably safer than live attenuated vaccines, especially for immunocompromised individuals.13 Current polio vaccines in Canada and the United States, for example, are inactivated instead of live attenuated,14 to eliminate disease risk due to inadequate attenuation. However, the downside of killed vaccines is that they may be less effective than live vaccines. For example, live attenuated vaccines for viruses and invasive bacteria can maintain their ability to access the cytosol, stimulating CD8+ T-cell responses against intracellular antigens,15 whereas killed vaccines elicit CD8+ T-cell immunity only via crosspresentation.15 In vaccination settings for which CD8+ T-cell responses are important, this may be a critical distinction.

Subunit vaccines deploy specific components of an agent rather than the whole organism. Subunit vaccines may use protein purified from the agent or recombinantly produced protein as antigens. Subunit vaccines often lack pathogen-associated molecular patterns, and thus typically cannot sufficiently initiate innate immune responses that drive the development of protection.10 Therefore, adjuvants (which stimulate stronger immune responses to antigens) are required for subunit vaccines. Adjuvants, the most common of which are alum salts, enhance innate immune engagement through sustained release and a variety of other mechanisms, including Toll-like receptor 4 (TLR4) stimulation, cytokine/chemokine production, and immune cell recruitment.16 Subunit vaccines can comprise different types of subunits. For example, toxoid vaccines are subunit vaccines that use as antigen an inactivated form of an otherwise disease-causing microbial toxin, such as tetanus toxin or diphtheria toxin.13 Other subunit vaccines, such as HPV vaccines, are engineered to form self-assembling virus-like particles.17 A polysaccharide can also serve as the subunit; however, polysaccharides are poorly immunogenic on their own13 and are, therefore, most often formulated as protein-polysaccharide conjugate vaccines, in which the protein component generates T-cell help to promote antipolysaccharide B-cell responses.13 Finally, viral vector and nucleic acid vaccines are subunit vaccine modalities that facilitate the translation of protein immunogens by host cells, thus eliciting immunity to this specific component of an agent.

Viral vector vaccines are engineered viruses that exploit viral host cell entry to deliver nucleic acids that encode for an antigen of interest. Viral vectors used for vaccination are typically highly attenuated or replication-incompetent to maximize safety.18,19 In addition, some viral vector platforms, such as lentivirus, can integrate into the genome and therefore pose a risk of insertional mutagenesis.20 Integration-defective lentiviral vectors, which are maintained as an episome, circumvent this issue.19 Importantly, preexisting immunity to the virus type used can hamper viral vector vaccine immunogenicity. Higher doses and/or additional doses can be attempted (which risks exacerbating the problem), or viral vector types with lower seroprevalence may be used.18 Furthermore, heterologous prime-boost strategies can be used to circumvent antivector immunity induced by the prime dose.18 

Nucleic acid vaccines contain DNA or messenger RNA (mRNA) encoding an antigen of interest, and must deliver their DNA or mRNA cargo into host cells. Encapsulation of nucleic acids in lipid nanoparticles (LNPs) enables effective nucleic acid delivery, as evidenced by severe acute respiratory syndrome coronavirus 2 mRNA LNP vaccines (mRNA-1273 and BNT162b2). LNP-based nucleic acid vaccines require cell uptake by endocytosis and endosomal escape to deliver their cargo.21 This is initiated by the ionizable lipid of LNP formulations, which becomes protonated under the low pH during endosome maturation, and leads to fusion of the LNP and endosomal membranes, releasing the cargo into the cytoplasm.21,22 In the case of mRNA vaccines, mRNA cargo can be directly translated in the cytoplasm following delivery. Alternatively, in the case of DNA vaccines, DNA cargo must reach the nucleus for transcription.23 This can be facilitated by nuclear localization signals or other mechanisms;23 however, DNA vaccines have shown limited success compared with mRNA vaccines, potentially due to this additional requirement for efficient antigen expression. mRNA used in nucleic acid vaccines requires several design considerations to optimize antigen expression, particularly involving capping, 5ʹ and 3ʹ untranslated region (UTR) sequences, and the poly-A tail.24 Furthermore, mRNA in these vaccines typically have modified nucleotides incorporated, such as N1-methylpseudouridine, which can improve immunogenicity by decreasing innate intracellular sensing that inhibits antigen translation.25-27 

Viral vector vaccines and LNP-encapsulated vaccines stimulate innate immunity and are, therefore, “self-adjuvanting,” avoiding the need for additional adjuvants in their formulation.18,28,29 These modalities, however, may be limited in applicability for some bacterial antigen targets. For example, data suggest mammalian-derived expression (such as that of viral vector and nucleic acid vaccines) of a bacterial antigen can result in limited native antigen recognition due to nonnative glycosylation and/or folding of bacterial antigen.30,31 This is in contrast to mammalian expression of viral antigen targets, such as severe acute respiratory syndrome coronavirus 2 Spike mRNA LNP vaccines, which would be expected to exhibit native glycosylation and structure, because viruses naturally rely on host machinery to propagate.

A major advantage of viral vector and nucleic acid vaccines is their ability to be rapidly developed using a “pipeline” approach, in which any antigen of interest may be fed into the manufacturing platform once its sequence is identified. The benefit of rapid development is well exemplified by the recent COVID-19 pandemic, for which the Moderna mRNA-1273 mRNA LNP vaccine went from sequence to clinical trial injections in only 66 days.32 

Lastly, vaccines may be monovalent (protecting against a single strain or pathogen) or multivalent (protecting against multiple strains or pathogens, also called combination vaccines). For example, current HPV vaccines are multivalent, designed for protection against multiple HPV strains (ie, quadrivalent Gardasil targeting HPV6/11/16/18).33 MMR vaccines are also multivalent, protecting against measles, mumps, and rubella.

Antigen selection

Antigen selection is a critical aspect of effective vaccine design. For live attenuated vaccines and inactivated/killed vaccines, where the pathogen itself defines the vaccine antigen content, representation of circulating variants of that pathogen is important. For example, each year, based on surveillance data, the World Health Organization selects the influenza strains predicted to be most prevalent for vaccine formulation, thereby maximizing overall protection.34 It can also be advantageous for the vaccine to represent the most infectious stage of a pathogen’s replication cycle. For example, live attenuated vaccines for malaria have been developed using Plasmodium falciparum sporozoites with promising initial results.35 Although the diverse antigen content delivered by whole-organism vaccines can provide broad coverage, a potential drawback is that responses may be primarily directed toward antigens that are poorly suited for establishing protection. For example, H pylori undergoes autolysis, releasing immunogenic intracellular proteins that may act as “decoys” and diminish responses to surface antigens.36 Additionally, immunodominant antigens may not be well conserved between strains, which can be a major barrier to effective protection for targets with significant strain diversity, such as Fusobacterium nucleatum.

Subunit vaccines allow rational selection of antigen target(s). However, antigen selection must be made with careful consideration of several factors. First, the antigen should be expressed on the surface of the agent, to be accessible to antibodies; second, the antigen should be conserved among the agents being targeted; third, the antigen should be consistently expressed, particularly during relevant phases of infection, such that the target is present/sufficiently abundant for targeting; fourth, the antigen should be specific to the pathogenic targets of interest, to minimize off-target effects (eg, disturbing commensal microbe populations and leading to dysbiosis) and, finally, the antigen should be functionally important to allow protection mediated by antibody-mediated neutralization. Indeed, many virus-targeted vaccines are protective via neutralization of proteins that enable infection.37-39 In the case of bacteria, neutralization can also provide protection through inhibition of key pathogenic functions of the antigen target.

Antigen avidity (the cumulative affinity achieved through multiple interactions) significantly affects immunogenicity following vaccination, with increased valency associated with greater B-cell activation and antibody titers.40,41 Recombinant subunit vaccines are advantageous in this regard as they support inclusion of multimerization domains, enabling multimeric antigen generation. For example, the multimeric nature of HPV virus-like particles is proposed as a primary reason for their exceptional potency.17 For viral vector and nucleic acid vaccines, surface display of antigen can effectively increase avidity by antigen localization/concentration on the cell surface, and has been shown to enhance antibody titers by approximately an order of magnitude compared with secreted monomeric antigen.42 

Timing of vaccination

Infectious agent vaccines are often effective prophylactics but generally ineffective therapeutics. Therapeutic vaccination is typically only viable in rare circumstances of long pathogen incubation periods (weeks to months), such as for rabies and HBV, during which vaccine-induced immunity can be established before widespread infection and acute disease. Aside from these exceptions, it is necessary that vaccination is timed to establish immunity before exposure to the microbe of interest. Therefore, understanding transmission underpins ideal vaccine timing for successful protection.

Booster vaccinations are also relevant to vaccine timing, and can be applied in 2 distinct manners. Firstly, boosters can be used to increase immunity above protective thresholds in cases where a single immunization elicits insufficient immunity. In this scenario, booster-induced immunity must therefore be established before exposure, as previously described. Alternatively, boosters can be used to maintain immunity above protective thresholds after waning. Timing of these boosters must be appropriately spaced to ensure constant levels above protective thresholds. For example, infants have generally weaker and shorter-lived immune responses to vaccination. Thus, booster series (often ≥2 vaccinations) are commonly used from infancy to early childhood to establish and maintain protective levels of immunity.43 

Route of administration

Vaccines can be administered through different routes, such as intramuscular, intranasal, IV, intradermal, subcutaneous, or oral. Route of administration can dictate the immune cell populations engaged and thus impact protection. Ideally, the route of vaccine delivery mimics the natural route of infection.15 For example, a major advantage of oral poliovirus vaccination is that oral administration can stimulate better mucosal immunity than intramuscular inactivated poliovirus vaccination, which may contribute to its greater protection against viral shedding.44,45 Similarly, it has been demonstrated that IV administration of malaria vaccines induces greater CD8+ and CD4+ T-cell responses in both peripheral blood and liver, and better protection compared with subcutaneous administration.35,46 

Vaccinating against oncomicrobes as a means of cancer control has long been a major interest of the scientific community, particularly for oncomicrobes with significant burden (Table 1). In this subsection, the successes, remaining challenges, and future directions of such efforts are discussed.

Successful oncomicrobe vaccines: HBV and HPV

HBV was the first oncomicrobe for which an anticancer vaccine was successfully developed. Chronic HBV infection is estimated to be responsible for 50% to 80% of hepatocellular carcinoma (HCC),57 the dominant form of primary liver cancer (∼80%).58 Infants perinatally infected are more susceptible to developing chronic HBV infection (80%-90% of infections become chronic) than children infected <6 years of age (20%-30%) or healthy individuals infected as adults (<5%),59 thus it is important to establish immunity shortly after birth.60 Accordingly, HBV vaccination consists of multiple doses given within the first 6 months.

Plasma-derived, first-generation HBV vaccines protect against infection with high efficacy.61 However, the long interval between HBV infection and HCC manifestation has made direct evaluation of protection against HCC difficult until recently. The first mass HBV vaccination program for infants began in 1984 in Taiwan,62,63 and reports comparing incidence before and after vaccine program implementation have shown early indications of protection.60 Perhaps the best current example is a randomized case-control study of infants vaccinated between 1983 and 1990 in China, which demonstrated protective efficacy of 72% and 70% against liver cancer incidence and mortality, respectively.48 Continued monitoring of these cohorts will be needed to evaluate protection into older age, as well as confirmation of similar results for modern recombinant HBV vaccine formulations; however, based on data available to date, HBV vaccination has the potential to dramatically decrease liver cancer incidence worldwide.

HPV is responsible for almost all (>99%) cervical cancers. Moreover, before HPV vaccine rollout, it was estimated that >90% of men and >80% of women, with ≥1 opposite sex partner, will acquire ≥1 HPV infection in their lifetime.64 Although most HPV infections spontaneously resolve (67% within 12 months), chronic infection is associated with greater risk of cervical cancer.65,66 Risk varies with HPV subtype, for example, HPV subtypes 16 and 18 are considered high risk for cervical cancer, whereas HPV subtypes 6 and 11 are considered low risk for cervical cancer (though are associated with ∼90% of genital warts).67,68 Because HPV is sexually transmitted, vaccination programs can be successfully applied prophylactically, before onset of sexual activity. Given the association of HPV with cervical cancer, initial HPV vaccination programs focused on vaccinating girls aged 9 to 14 years. However, HPV is also associated with lower-incident cancers, such as oral cavity, penile, anal, vulvar, vaginal, oropharyngeal, and laryngeal.4 Therefore, immunization of boys is now recommended as a secondary target group.69 These vaccinations should also increase herd effects and further benefit girls as well.

As with HBV vaccination, time was required for the effects of HPV vaccination on cancer incidence to become evaluable. Recent assessments indicate an 88% reduction in cervical cancer incidence in adolescent girls vaccinated with quadrivalent HPV vaccines before 17 years of age.47 The continued effect of HPV vaccines on reduction of cervical cancer incidence over time, as these cohorts age, and the anticipated protection against other HPV-associated cancers, will indicate the full potential of these vaccines. Although HPV16 and HPV18 are covered by all HPV vaccines, and were initially prioritized in bivalent and quadrivalent designs on the basis they account for ∼70% of total cervical cancers,47 the more recent 9-valent HPV vaccination programs protect against additional high-risk cancer-related HPV strains,70 and are expected to further reduce the incidence of HPV-associated cancers.

Ongoing challenges: H pylori and EBV

In the 1980s, Barry Marshall famously drank a culture of H pylori and developed gastritis, supporting, anecdotally, a causative relationship between H pylori infection and gastric inflammation that has since been verified.71 Over 40% of the global adult population is estimated to be infected with H pylori, though there is extensive geographic variation in carriage rates.72 Infection is believed to predominantly occur during childhood.73 Although some individuals can clear H pylori infection without treatment, as was the case for Barry Marshall,74 most infections persist for life unless treated.73H pylori can be routinely detected (eg, by urea breath test) and treated with antibiotics.75 However, most infections are asymptomatic,73 and therefore many infected patients do not seek treatment and remain chronically infected. Concerningly, a high prevalence of pretreatment isolates (≥10%) show resistance to common antibiotics.76 Additionally, high rates of reoccurrence, either through reinfection (particularly in developing countries with higher prevalence) or through recrudescence (ie, incomplete elimination), further highlight that antibiotics have limited effectiveness in disease control, and emphasizes the need for alternative interventions that confer long-lived immune memory and protection.77,78 

H pylori has been estimated to be associated with 75% to 80% of gastric cancers.4H pylori can survive the acidic stomach lumen during colonization due, in part, to its production of urease, which converts urea to ammonia,79 neutralizing stomach acid and creating a more favorable environment for the microbe. Further, H pylori is highly motile, and can embed itself in the gastric mucosa, adhere to epithelial cells, and exhibits various immune evasion mechanisms that support its long-term persistence in the stomach.79 

H pylori elimination can improve cancer-related outcomes. For example, a recent systematic review of H pylori eradication therapy reported a relative risk of 0.54 for gastric cancer incidence in treated healthy individuals compared with placebo or no treatment healthy control subjects.80 However, given the widespread prevalence and often asymptomatic nature of H pylori infection, and the potential for further emergence of antibiotic-resistance strains, early childhood vaccination has the best potential for significant reduction of gastric cancer incidence. Unfortunately, despite the now long-standing recognition of the role of H pylori in gastric cancer, and the significant effort toward developing such vaccines, an effective, approved, H pylori vaccine has remained elusive due to immune evasion mechanisms, significant genetic diversity, and, as mice are not natural H pylori hosts, model systems that do not fully emulate human infection.81 Nevertheless, some H pylori vaccine clinical trials have shown promise. For example, an orally administered H pylori vaccine using a fusion protein of urease B subunit of H pylori and heat-labile enterotoxin B subunit of Escherichia coli, the latter of which acts as a mucosal adjuvant, was evaluated in a phase 3 clinical trial.82 This strategy demonstrated 71.8%, 55.0%, and 55.8% efficacy against H pylori infection in naïve individuals at 12, 24, and 36 months, respectively.82 Unfortunately, however, the vaccine has since been discontinued,83 and therefore additional investigations of H pylori vaccine approaches, ideally with greater long-term protection, are needed.

EBV has been found to infect >90% of the global population,84 establishing a life-long latency. EBV has been linked to numerous conditions, including infectious mononucleosis (IM), nasopharyngeal cancers (85% of incidences), gastric cancer (7.7% of incidences), and Hodgkin (50% of incidences) and non-Hodgkin lymphomas (55% of incidences),4,49,85 including Burkitt lymphoma.86 

In the 1970s, Anthony Epstein proposed the development of an EBV vaccine to reduce the burden of EBV-associated cancers.87 Since then, most vaccine development efforts have focused on the most abundant glycoprotein (gp) expressed on EBV’s envelope, gp350.88 An adjuvanted gp350 monomer progressed to phase 2 clinical trials and demonstrated 98.7% seroconversion in vaccinated participants. Although EBV infection was not prevented, rates of IM were reduced.89,90 This is an important finding, as an EBV vaccine that controls viral load may protect against associated cancers even without achieving sterilizing-immunity.84 No further reports on this vaccine have been made. However, a gp350-ferritin nanoparticle vaccine, which elicited robust neutralizing antibody responses in mice and nonhuman primates, and demonstrated efficacy in a recombinant virus challenge murine model,91 is now in phase 1 clinical testing (ClinicalTrials.gov identifier: NCT04645147).92 Other EBV vaccines have targeted the membrane fusion molecules gB and gH/gL.88 In rabbits vaccinated with trimeric or monomeric gH/gL, trimeric gB, and tetrameric gp350, higher neutralizing titers were observed relative to monomeric gp350.93 Additionally, Moderna has launched a phase 1 trial for a multivalent mRNA vaccine encoding gH, gL, gp42, and gp220 to target IM (NCT05164094),88 and a therapeutic candidate for long-term sequalae (NCT05831111) and, more recently, a phase 1 study evaluating an adjuvanted gH/gL/gp42-ferritin nanoparticle vaccine has been launched (NCT06908096).

As emphasized by the >10 years of development between the classification of HPV as a group 1 carcinogen and approvals for HPV vaccinations, and the lack of approved H pylori vaccines, now at >30 years from its classification as a group 1 carcinogen,82 vaccine development requires considerable research investment toward identifying underlying features of pathogenesis and protective immunity, and efficacious formulations. Nevertheless, as now evidenced by HBV and HPV vaccines, successful development of such vaccines can have significant impacts on reducing cancer incidence and, therefore, continued efforts are needed.

Vaccine development against emerging oncomicrobes: ETBF, pks+E coli, and F nucleatum

In recent years, enterotoxigenic Bacteroides fragilis (ETBF), polyketide synthase–positive (pks+) E coli, and F nucleatum have emerged as novel oncomicrobes in the setting of colorectal cancer (CRC). In this subsection, the biology and roles of ETBF, pks+E coli, and F nucleatum in cancer are discussed, and potential strategies for vaccine development against these agents are proposed.

B fragilis is a normal inhabitant of the healthy human colon. Two subtypes exist: nontoxigenic B fragilis (NTBF) and ETBF, and it is specifically the ETBF subtype that is enriched in sporadic and hereditary CRC.94,95 Further, ETBF, but not NTBF, has been shown to potentiate the development of colonic tumors in APCmin/+ mouse models.96 The distinguishing feature between these 2 subtypes is BFT, a 20-kilodalton toxin present in ETBF and absent in NTBF. BFT cleaves E-cadherin,97 stimulating β-catenin signaling and cell proliferation,98 and can disrupt epithelial cell barriers,99,100 and lead to recruitment of myeloid cells to the distal colon, which contributes to tumorigenesis in murine models.101 As the procarcinogenic action of ETBF can be directly attributed to BFT, a toxoid vaccine deploying a mutated, recombinant variant of the toxin that lacks toxin functionality but elicits antitoxin immunity has potential. Indeed, targeted mutagenesis to ablate E-cadherin cleavage has been previously determined.102 

E coli variants bearing a 50-kilobase pathogenicity island encoding a pks103 are enriched in tumor specimens of spontaneous and hereditary CRC.95,104 The product of this pks, colibactin, is genotoxic, causing DNA double-strand breaks, interstrand crosslinks, and chromosomal aberrations.105-107 Intestinal organoids repeatedly exposed to pks+E coli have been used to identify 2 colibactin-specific mutational signatures.103 These signatures, either individually or in combination, were found to be present in 2% to 8.8% of primary and metastatic CRC samples. Moreover, the colibactin target motif represented 2.4% of driver mutations in CRC, supporting a direct initiating role of colibactin in at least a small fraction of CRC.103 

Although colibactin is the direct mediator of this mutational signature and therefore would be an ideal vaccine target in the same manner as BFT, colibactin, as a secondary metabolite, is poorly immunogenic, which limits the potential of this strategy. A protein-conjugate vaccine approach could possibly be explored in this scenario; however, colibactin is also highly labile, which may make protein-conjugate vaccine development challenging. Recent data showing epithelial cell adhesion is required for colibactin’s mutational effects provide another potential tactic.108 The 2 adhesins responsible for binding of E coli to epithelial cells, FimH and FmlH, may be effective vaccine targets, through which blockade of adhesion may prevent the localized production of colibactin that enables DNA damage by this rapidly destabilized mutagen. Consistent with this strategy, a FimH-neutralizing small molecule, siobofimloc, was effective in reducing DNA damage and tumor burden in pks+E coli–exposed mice.108 FimH vaccines have been developed and shown to be effective for adhesion blockade of E coli in other contexts, such as urinary tract infections, and may be evaluated for benefit in CRC as well.109,110 Given the pks mutational signature is established before 10 years of age,103,111 such a vaccine would need to be given in the first years of life to act prophylactically.

Given the colonic niche of both BFT and pks+E coli, vaccines inducing strong mucosal responses in the gastrointestinal tract would be most applicable to protection. An orally administered vaccine may therefore be appropriate. However, both situations require use of specific subunit antigens to prevent targeting of related commensals (ie, NTBF, pksE coli), and subunit vaccines have significant barriers to oral applications, such as stability during transit through the stomach and overcoming tolerogenic mechanisms.112 Indeed, all licensed oral vaccines are formulated with live attenuated or killed whole cells.113 Furthermore, nonlive attenuated mucosal vaccines can suffer from short-lived protection, particularly in children (which would be the ideal target population for ETBF and BFT vaccination due to early colonization by these oncomicrobes). The most relevant example is Dukoral, comprised of inactivated Vibrio cholera and recombinant cholera toxin B subunit. Protection against cholera conferred by this vaccine is lower and rapidly wanes in children aged 2-5 years compared with individuals aged >5 years.114 Furthermore, even protection in individuals aged >5 years is relatively short-lived (decreasing from 78% to 40% after the first and third year, respectively),114 compared with decade or lifetime protection conferred by many systemic vaccines. Thus, novel strategies may need to be developed and used for effective, long-lived mucosal immunity in the context of BFT and pks+E coli vaccination.

F nucleatum is a gram-negative, anaerobic bacterium ubiquitous in the oral cavity, and acts largely as a commensal microbe in this setting. Oral bacteria such as F nucleatum can escape the oral cavity during transient, low-level bacteremia that commonly occurs from everyday activities such as toothbrushing.115 When F nucleatum spreads hematogenously, its adhesive properties allow it to selectively colonize peripheral niches including tumors, mainly CRC,116,117 but also breast,53 esophageal,54 pancreatic,118 oral/head and neck,55 and cervical cancers.119 Tumor colonization by F nucleatum is mediated by its 2 key adhesin proteins, Fap2 and RadD, which bind to GalGalNAc and CD147, respectively, which are both overexpressed on the surface of some tumor cell types.120,121 

F nucleatum is distinct from the other oncomicrobes in this review in being a cancer promoter rather than a cancer initiator. Nonetheless, associations of F nucleatum with increased tumor growth,53,122 chemoresistance,123 decreased effectiveness of immunotherapy,124,125 metastasis,126 recurrence,127 and poor patient outcomes128 are sufficient to prompt vaccine development efforts. F nucleatum, however, is a particularly difficult vaccine target, due to the need to continuously protect against systemic dissemination from the oral cavity, and its tropism for tumors, which are immunosuppressive microenvironments and therefore can offer safe harbor from host immunity. This requires a F nucleatum vaccine to induce sufficiently robust circulating serum antibodies to intercept bloodborne F nucleatum before tumor colonization. Toward this goal, IV vaccination may be well suited for F nucleatum, for the reasons described above for malaria IV vaccination.

Another major challenge to vaccinating against F nucleatum is the extraordinary strain diversity of this bacterium. Early experimentation with whole-cell vaccine approaches for F nucleatum demonstrated poor cross-reactivity, with >60% of antibodies isolated following whole-cell vaccination recognizing only a single isolate.129 Subunit vaccines targeting conserved regions of the adhesins Fap2 and RadD may be a promising alternative strategy to prevent F nucleatum tumor colonization, analogous to targeting FimH/FmlH to disrupt the adherent properties of pks+E coli.

For oncomicrobes, vaccination timing can be prophylactic relative to infection or relative to tumor development. Prophylactic vaccination against ETBF and pks+E coli before tumor development, and ideally in early life, is well justified, because these agents are confirmed tumor initiators. This could be especially beneficial for individuals with elevated CRC risk, such as patients with ulcerative colitis (ie, 2%, 8%, or 18% of patients with ulcerative colitis develop CRC after 10, 20, or 30 years with the disease, respectively).130 Alternatively, the aim of vaccinating against F nucleatum would be to prevent tumor promotion by this agent by blocking its spread from the commensal oral environment to an ectopic tumor environment. In addition, F nucleatum vaccination could be coordinated with standard radiotherapy and chemotherapy approaches, to prevent recolonization upon relapse, or coordinated with antibiotic therapy, to prevent recolonization after a course of antibiotics has eliminated the original bacterial population from sites of minimal residual disease.

It is also important to highlight that reduction of cancer incidence is not a viable end point for clinical trials that would lead to initial oncomicrobe vaccine approvals, due to the long delay between infection and cancer manifestation. For example, HBV vaccines were approved on the basis of effective prevention of acute hepatitis, not HCC.61 Similarly, HPV vaccines were approved based on reduction of precancerous cervical lesions, as a surrogate end point for cervical cancer.131 For H pylori and EBV, primary outcomes of prevention of gastritis and IM, respectively, could be used. Clinical evaluation of F nucleatum vaccination in combination scenarios previously described may provide a path to initial approval. Monitoring rates and outcomes of subsequent cancers in these cohorts may justify more widespread applications. Alternatively, BFT vaccination could be evaluated for prevention of diarrheal diseases, for which its association is well established.132 For pks+E coli, prevention of infection, and specifically prevention of its related mutational signature, which is established early in life, may be an ideal outcome of interest. Such preliminary data from these vaccines would be informative with respect to potential for cancer control. Given the large number of individuals that would stand to benefit from protection against these novel oncomicrobes, the potential of ETBF, pks+E coli, and F nucleatum vaccination demands investigation.

We are in an exciting era in which the cancer-reducing potential of HBV and HPV vaccination is being substantiated by population-scale, real-world data, validating vaccination as a viable means of controlling infectious agent-associated cancers. It is our sincere hope that such results will reinvigorate the development of vaccines against both established and novel oncomicrobial targets.

This work was supported by Cancer Research UK (C54768/A29062). This work was also supported by the Canadian Glycomics Network (GlycoNet), a member of the Networks of Centres of Excellence Canada program. C.A.D. was supported by Canadian graduate scholarships from the Canadian Institutes of Health Research.

Contribution: C.A.D., N.J.P.V., and R.A.H. wrote the manuscript.

Conflict-of-interest disclosure: C.A.D., N.J.P.V., and R.A.H. are working on developing Fusobacterium nucleatum vaccines with potential for institutional patent filings.

Correspondence: Robert A. Holt, British Columbia Cancer Research Institute, 675 W 10th Ave, Vancouver, BC V5Z 1L3, Canada; email: rholt@bcgsc.ca.

1.
Carter
A
,
Msemburi
W
,
Sim
SY
, et al
.
Modeling the impact of vaccination for the immunization Agenda 2030: deaths averted due to vaccination against 14 pathogens in 194 countries from 2021 to 2030
.
Vaccine
.
2024
;
42
(
suppl 1
):
S28
-
S37
.
2.
Rous
P
.
A sarcoma of the fowl transmissible by an agent separable from the tumor cells
.
J Exp Med
.
1911
;
13
(
4
):
397
-
411
.
3.
Elemento
O
.
The road from Rous sarcoma virus to precision medicine
.
J Exp Med
.
2021
;
218
(
4
):
e20201754
.
4.
de Martel
C
,
Georges
D
,
Bray
F
,
Ferlay
J
,
Clifford
GM
.
Global burden of cancer attributable to infections in 2018: a worldwide incidence analysis
.
Lancet Glob Health
.
2020
;
8
(
2
):
e180
-
e190
.
5.
Schiller
JT
,
Lowy
DR
.
Prospects for preventing cancer with anti-microbial prophylactic vaccines
.
Cell Host Microbe
.
2023
;
31
(
1
):
137
-
140
.
6.
Bouvard
V
,
Baan
R
,
Straif
K
, et al
.
A review of human carcinogens--part B: biological agents
.
Lancet Oncol
.
2009
;
10
(
4
):
321
-
322
.
7.
Fine
P
,
Eames
K
,
Heymann
DL
.
“Herd immunity”: a rough guide
.
Clin Infect Dis
.
2011
;
52
(
7
):
911
-
916
.
8.
John
TJ
,
Samuel
R
.
Herd immunity and herd effect: new insights and definitions
.
Eur J Epidemiol
.
2000
;
16
(
7
):
601
-
606
.
9.
Parrino
J
,
Graham
BS
.
Smallpox vaccines: past, present, and future
.
J Allergy Clin Immunol
.
2006
;
118
(
6
):
1320
-
1326
.
10.
Di Pasquale
A
,
Preiss
S
,
Tavares Da Silva
F
,
Garçon
N
.
Vaccine adjuvants: from 1920 to 2015 and beyond
.
Vaccines (Basel)
.
2015
;
3
(
2
):
320
-
343
.
11.
Plotkin
S
.
History of vaccination
.
Proc Natl Acad Sci U S A
.
2014
;
111
(
34
):
12283
-
12287
.
12.
Hsu
T
,
Hingley-Wilson
SM
,
Chen
B
, et al
.
The primary mechanism of attenuation of bacillus Calmette-Guérin is a loss of secreted lytic function required for invasion of lung interstitial tissue
.
PNAS
.
2003
;
100
(
21
):
12420
-
12425
.
13.
Pollard
AJ
,
Bijker
EM
.
A guide to vaccinology: from basic principles to new developments
.
Nat Rev Immunol
.
2021
;
21
(
2
):
83
-
100
.
14.
Alfaro-Murillo
JA
,
Ávila-Agüero
ML
,
Fitzpatrick
MC
,
Crystal
CJ
,
Falleiros-Arlant
LH
,
Galvani
AP
.
The case for replacing live oral polio vaccine with inactivated vaccine in the Americas
.
The Lancet
.
2020
;
395
(
10230
):
1163
-
1166
.
15.
Murphy
K
,
Weaver
C
,
Berg
L
. Janeway’s Immunobiology. (10th ed) .
W.W. Norton and Company
;
2022
.
16.
Pulendran
B
,
S Arunachalam
P
,
O’Hagan
DT
.
Emerging concepts in the science of vaccine adjuvants
.
Nat Rev Drug Discov
.
2021
;
20
(
6
):
454
-
475
.
17.
Schiller
J
,
Lowy
D
.
Explanations for the high potency of HPV prophylactic vaccines
.
Vaccine
.
2018
;
36
(
32 pt A
):
4768
-
4773
.
18.
McCann
N
,
O’Connor
D
,
Lambe
T
,
Pollard
AJ
.
Viral vector vaccines
.
Curr Opin Immunol
.
2022
;
77
:
102210
.
19.
Travieso
T
,
Li
J
,
Mahesh
S
,
Mello
JDFRE
,
Blasi
M
.
The use of viral vectors in vaccine development
.
NPJ Vaccin
.
2022
;
7
(
1
):
75
.
20.
Howe
SJ
,
Mansour
MR
,
Schwarzwaelder
K
, et al
.
Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients
.
J Clin Invest
.
2008
;
118
(
9
):
3143
-
3150
.
21.
Chatterjee
S
,
Kon
E
,
Sharma
P
,
Peer
D
.
Endosomal escape: a bottleneck for LNP-mediated therapeutics
.
Proc Natl Acad Sci U S A
.
2024
;
121
(
11
):
e2307800120
.
22.
Cullis
PR
,
Felgner
PL
.
The 60-year evolution of lipid nanoparticles for nucleic acid delivery
.
Nat Rev Drug Discov
.
2024
;
23
(
9
):
709
-
722
.
23.
Shafaati
M
,
Saidijam
M
,
Soleimani
M
, et al
.
A brief review on DNA vaccines in the era of COVID-19
.
Future Virol
.
2021
;
17
(
1
):
49
-
66
.
24.
Kon
E
,
Elia
U
,
Peer
D
.
Principles for designing an optimal mRNA lipid nanoparticle vaccine
.
Curr Opin Biotechnol
.
2022
;
73
:
329
-
336
.
25.
Karikó
K
,
Muramatsu
H
,
Ludwig
J
,
Weissman
D
.
Generating the optimal mRNA for therapy: HPLC purification eliminates immune activation and improves translation of nucleoside-modified, protein-encoding mRNA
.
Nucleic Acids Res
.
2011
;
39
(
21
):
e142
.
26.
Pardi
N
,
Hogan
MJ
,
Porter
FW
,
Weissman
D
.
mRNA vaccines-a new era in vaccinology
.
Nat Rev Drug Discov
.
2018
;
17
(
4
):
261
-
279
.
27.
Bernard
MC
,
Bazin
E
,
Petiot
N
, et al
.
The impact of nucleoside base modification in mRNA vaccine is influenced by the chemistry of its lipid nanoparticle delivery system
.
Mol Ther Nucleic Acids
.
2023
;
32
:
794
-
806
.
28.
Wang
S
,
Liang
B
,
Wang
W
, et al
.
Viral vectored vaccines: design, development, preventive and therapeutic applications in human diseases
.
Signal Transduct Target Ther
.
2023
;
8
(
1
):
149
.
29.
Connors
J
,
Joyner
D
,
Mege
NJ
, et al
.
Lipid nanoparticles (LNP) induce activation and maturation of antigen presenting cells in young and aged individuals
.
Commun Biol
.
2023
;
6
(
1
):
188
.
30.
Ozdilek
A
,
Paschall
AV
,
Dookwah
M
,
Tiemeyer
M
,
Avci
FY
.
Host protein glycosylation in nucleic acid vaccines as a potential hurdle in vaccine design for nonviral pathogens
.
Proc Natl Acad Sci U S A
.
2020
;
117
(
3
):
1280
-
1282
.
31.
Ozdilek
A
,
Avci
FY
.
Glycosylation as a key parameter in the design of nucleic acid vaccines
.
Curr Opin Struct Biol
.
2022
;
73
:
102348
.
32.
Jackson
LA
,
Anderson
EJ
,
Rouphael
NG
, et al
.
An mRNA vaccine against SARS-CoV-2 — preliminary report
.
N Engl J Med
.
2020
;
383
(
20
):
1920
-
1931
.
33.
Soliman
M
,
Oredein
O
,
Dass
CR
.
Update on safety and efficacy of HPV vaccines: focus on Gardasil
.
Int J Mol Cell Med
.
2021
;
10
(
2
):
101
-
113
.
34.
CDC
.
Selecting Viruses for the Seasonal Influenza Vaccine
. Accessed 10 October 2025. https://www.cdc.gov/flu/vaccine-process/vaccine-selection.html.
35.
Richie
TL
,
Church
LWP
,
Murshedkar
T
, et al
.
Sporozoite immunization: innovative translational science to support the fight against malaria
.
Expert Rev Vaccin
.
2023
;
22
(
1
):
964
-
1007
.
36.
Phadnis
SH
,
Parlow
MH
,
Levy
M
, et al
.
Surface localization of Helicobacter pylori urease and a heat shock protein homolog requires bacterial autolysis
.
Infect Immun
.
1996
;
64
(
3
):
905
-
912
.
37.
Plotkin
SA
.
Immunologic correlates of protection induced by vaccination
.
Pediatr Infect Dis J
.
2001
;
20
(
1
):
63
-
75
.
38.
Plotkin
SA
.
Correlates of vaccine-induced immunity
.
Clin Infect Dis
.
2008
;
47
(
3
):
401
-
409
.
39.
Plotkin
SA
.
Correlates of protection induced by vaccination
.
Clin Vaccin Immunol
.
2010
;
17
(
7
):
1055
-
1065
.
40.
Kato
Y
,
Abbott
RK
,
Freeman
BL
, et al
.
Multifaceted effects of antigen valency on B cell response composition and differentiation in vivo
.
Immunity
.
2020
;
53
(
3
):
548
-
563.e8
.
41.
Lainšček
D
,
Fink
T
,
Forstnerič
V
, et al
.
A nanoscaffolded spike-RBD vaccine provides protection against SARS-CoV-2 with minimal anti-scaffold response
.
Vaccines (Basel)
.
2021
;
9
(
5
):
431
.
42.
Komori
M
,
Nogimori
T
,
Morey
AL
, et al
.
saRNA vaccine expressing membrane-anchored RBD elicits broad and durable immunity against SARS-CoV-2 variants of concern
.
Nat Commun
.
2023
;
14
(
1
):
2810
.
43.
Semmes
EC
,
Chen
JL
,
Goswami
R
,
Burt
TD
,
Permar
SR
,
Fouda
GG
.
Understanding early-life adaptive immunity to guide interventions for pediatric health
.
Front Immunol
.
2020
;
11
:
595297
.
44.
Connor
RI
,
Brickley
EB
,
Wieland-Alter
WF
, et al
.
Mucosal immunity to poliovirus
.
Mucosal Immunol
.
2022
;
15
(
1
):
1
-
9
.
45.
Hird
TR
,
Grassly
NC
.
Systematic review of mucosal immunity induced by oral and inactivated poliovirus vaccines against virus shedding following oral poliovirus challenge
.
Plos Pathog
.
2012
;
8
(
4
):
e1002599
.
46.
Epstein
JE
,
Tewari
K
,
Lyke
KE
, et al
.
Live attenuated malaria vaccine designed to protect through hepatic CD8 + T cell immunity
.
Science
.
1979
;
2011
:
334
.
47.
Lei
J
,
Ploner
A
,
Elfström
KM
, et al
.
HPV vaccination and the risk of invasive cervical cancer
.
N Engl J Med
.
2020
;
383
(
14
):
1340
-
1348
.
48.
Cao
M
,
Fan
J
,
Lu
L
, et al
.
Long term outcome of prevention of liver cancer by hepatitis B vaccine: results from an RCT with 37 years
.
Cancer Lett
.
2022
;
536
:
215652
.
49.
Camargo
MC
,
Murphy
G
,
Koriyama
C
, et al
.
Determinants of Epstein-Barr virus-positive gastric cancer: an international pooled analysis
.
Br J Cancer
.
2011
;
105
(
1
):
38
-
43
.
50.
Xia
S
,
Ma
L
,
Li
H
,
Li
Y
,
Yu
L
.
Prevalence of enterotoxigenic Bacteroides fragilis in patients with colorectal cancer: a systematic review and meta-analysis
.
Front Cell Infect Microbiol
.
2025
;
15
:
1525609
.
51.
de Souza
JB
,
de Almeida Campos
LA
,
Palácio
SB
,
Brelaz-de-Castro
MCA
,
Cavalcanti
IMF
.
Prevalence and implications of pKs-positive Escherichia coli in colorectal cancer
.
Life Sci
.
2024
;
341
:
122462
.
52.
Janati
AI
,
Karp
I
,
Laprise
C
,
Sabri
H
,
Emami
E
.
Detection of Fusobaterium nucleatum in feces and colorectal mucosa as a risk factor for colorectal cancer: a systematic review and meta-analysis
.
Syst Rev
.
2020
;
9
(
1
):
276
.
53.
Parhi
L
,
Alon-Maimon
T
,
Sol
A
, et al
.
Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression
.
Nat Commun
.
2020
;
11
(
1
):
3259
.
54.
Yamamura
K
,
Baba
Y
,
Nakagawa
S
, et al
.
Human microbiome Fusobacterium nucleatum in esophageal cancer tissue is associated with prognosis
.
Clin Cancer Res
.
2016
;
22
(
22
):
5574
-
5581
.
55.
Bronzato
JD
,
Bomfim
RA
,
Edwards
DH
,
Crouch
D
,
Hector
MP
,
Gomes
BPFA
.
Detection of Fusobacterium in oral and head and neck cancer samples: a systematic review and meta-analysis
.
Arch Oral Biol
.
2020
;
112
(
August 2019
). 104669-20.
56.
Bray
F
,
Ferlay
J
,
Soerjomataram
I
,
Siegel
RL
,
Torre
LA
,
Jemal
A
.
Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
.
2018
;
68
(
6
):
394
-
424
.
57.
Venook
AP
,
Papandreou
C
,
Furuse
J
,
de Guevara
LL
,
Guevara
DE
.
The incidence and epidemiology of hepatocellular carcinoma: a global and regional perspective
.
Oncologist
.
2010
;
15
(
suppl 4
):
5
-
13
.
58.
Rumgay
H
,
Ferlay
J
,
de Martel
C
, et al
.
Global, regional and national burden of primary liver cancer by subtype
.
Eur J Cancer
.
2022
;
161
:
108
-
118
.
59.
Hepatitis B Vaccines: WHO Position Paper – July 2017. Weekly Epidemiological Record
.
WHO
;
2017
. 92.
60.
Flores
JE
,
Thompson
AJ
,
Ryan
M
,
Howell
J
.
The global impact of hepatitis B vaccination on hepatocellular carcinoma
.
Vaccines (Basel)
.
2022
;
10
(
5
):
793
.
61.
Szmuness
W
,
Stevens
CE
,
Zang
EA
,
Harley
EJ
,
Kellner
A
.
A controlled clinical trial of the efficacy of the hepatitis B vaccine (heptavax B): a final report
.
Hepatology
.
1981
;
1
(
5
):
377
-
385
.
62.
Chang
MH
. Hepatitis B Virus and Cancer Prevention. In:
Senn
HJ
,
Otto
F
, eds.
Clinical Cancer Prevention. Recent Results in Cancer Research
.
Berlin, Heidelberg
:
Springer
;
2010
.
63.
Chen
DS
,
Hsu
NH
,
Sung
JL
, et al
.
A mass vaccination program in Taiwan against hepatitis B virus infection in infants of hepatitis B surface antigen-carrier mothers
.
JAMA
.
1987
;
257
(
19
):
2597
-
2603
.
64.
Chesson
HW
,
Dunne
EF
,
Hariri
S
,
Markowitz
LE
.
The estimated lifetime probability of acquiring human papillomavirus in the United States
.
Sex Transm Dis
.
2014
;
41
(
11
):
660
-
664
.
65.
Moscicki
AB
,
Schiffman
M
,
Franceschi
S
. The natural history of human papillomavirus infection in relation to cervical cancer. In:
Jenkins
D
,
Bosch
FX
, eds.
Human Papillomavirus: Proving and Using a Viral Cause for Cancer
.
Elsevier
;
2019
:
149
-
160
.
66.
Rodríguez
AC
,
Schiffman
M
,
Herrero
R
, et al
.
Rapid clearance of human papillomavirus and implications for clinical focus on persistent infections
.
J Natl Cancer Inst
.
2008
;
100
(
7
):
513
-
517
.
67.
Muñoz
N
,
Bosch
FX
,
De Sanjosé
S
, et al
.
Epidemiologic classification of human papillomavirus types associated with cervical cancer
.
N Engl J Med
.
2003
;
348
(
6
):
518
-
527
.
68.
Chelimo
C
,
Wouldes
TA
,
Cameron
LD
,
Elwood
JM
.
Risk factors for and prevention of human papillomaviruses (HPV), genital warts and cervical cancer
.
J Infect
.
2013
;
66
(
3
):
207
-
217
.
69.
World Health Organization
. Human Papillomavirus Vaccines: WHO Position Paper (2022 Update). Weekly Epidemiological Record.
WHO
;
2022
.
70.
Kim
J
,
Choe
YJ
,
Park
J
, et al
.
Comparative effects of bivalent, quadrivalent, and nonavalent human papillomavirus vaccines in the prevention of genotype-specific infection: a systematic review and network meta-analysis
.
Infect Chemother
.
2024
;
56
(
1
):
37
-
46
.
71.
Hellström
PM
.
This year’s Nobel Prize to gastroenterology: Robin Warren and Barry Marshall awarded for their discovery of Helicobacter pylori as pathogen in the gastrointestinal tract
.
World J Gastroenterol
.
2006
;
12
(
19
):
3126
.
72.
Li
Y
,
Choi
H
,
Leung
K
,
Jiang
F
,
Graham
DY
,
Leung
WK
.
Global prevalence of Helicobacter pylori infection between 1980 and 2022: a systematic review and meta-analysis
.
Lancet Gastroenterol Hepatol
.
2023
;
8
(
6
):
553
-
564
.
73.
Malfertheiner
P
,
Camargo
MC
,
El-Omar
E
, et al
.
Helicobacter pylori infection
.
Nat Rev Dis Primers
.
2023
;
9
(
1
):
19
.
74.
Marshall
B
. A Brief History of the Discovery of Helicobacter pylori. In:
Suzuki
H
,
Warren
R
,
Marshall
B
, eds.
Helicobacter pylori
.
Tokyo
:
Springer
;
2016
.
75.
Crowe
SE
.
Helicobacter pylori Infection
.
N Engl J Med
.
2019
;
380
(
12
):
1158
-
1165
.
76.
Savoldi
A
,
Carrara
E
,
Graham
DY
,
Conti
M
,
Tacconelli
E
.
Prevalence of antibiotic resistance in Helicobacter pylori: a systematic review and meta-analysis in World Health Organization regions
.
Gastroenterology
.
2018
;
155
(
5
):
1372
-
1382.e17
.
77.
Niv
Y
,
Hazazi
R
.
Helicobacter pylori recurrence in developed and developing countries: meta-analysis of 13C-urea breath test follow-up after eradication
.
Helicobacter
.
2008
;
13
(
1
):
56
-
61
.
78.
Hu
Y
,
Wan
JH
,
Li
XY
,
Zhu
Y
,
Graham
DY
,
Lu
NH
.
Systematic review with meta-analysis: the global recurrence rate of Helicobacter pylori
.
Aliment Pharmacol Ther
.
2017
;
46
(
9
):
773
-
779
.
79.
Fan
J
,
Zhu
J
,
Xu
H
.
Strategies of Helicobacter pylori in evading host innate and adaptive immunity: insights and prospects for therapeutic targeting
.
Front Cell Infect Microbiol
.
2024
;
14
:
1342913
.
80.
Ford
AC
,
Yuan
Y
,
Moayyedi
P
.
Helicobacter pylori eradication therapy to prevent gastric cancer: systematic review and meta-analysis
.
Gut
.
2020
;
69
(
12
):
2113
-
2121
.
81.
Li
S
,
Zhao
W
,
Xia
L
,
Kong
L
,
Yang
L
.
How long will it take to launch an effective Helicobacter pylori vaccine for humans?
.
Infect Drug Resist
.
2023
;
16
:
3787
-
3805
.
82.
Zeng
M
,
Mao
XH
,
Li
JX
, et al
.
Efficacy, safety, and immunogenicity of an oral recombinant Helicobacter pylori vaccine in children in China: a randomised, double-blind, placebo-controlled, phase 3 trial
.
The Lancet
.
2015
;
386
(
10002
):
1457
-
1464
.
83.
Sutton
P
,
Boag
JM
.
Status of vaccine research and development for Helicobacter pylori
.
Vaccine
.
2019
;
37
(
50
):
7295
-
7299
.
84.
Cohen
JI
,
Fauci
AS
,
Varmus
H
,
Nabel
GJ
.
Epstein-Barr virus: an important vaccine target for cancer prevention
.
Sci Transl Med
.
2011
;
3
(
107
):
107fs7
.
85.
Damania
B
,
Kenney
SC
,
Raab-Traub
N
.
Epstein-Barr virus: biology and clinical disease
.
Cell
.
2022
;
185
(
20
):
3652
-
3670
.
86.
Brady
G
,
MacArthur
GJ
,
Farrell
PJ
.
Epstein-Barr virus and Burkitt lymphoma
.
J Clin Pathol
.
2008
;
84
(
993
):
372
-
377
.
87.
Balfour
HH
.
Progress, prospects, and problems in Epstein-Barr virus vaccine development
.
Curr Opin Virol
.
2014
;
6
(
1
):
1
-
5
.
88.
Zhong
L
,
Krummenacher
C
,
Zhang
W
, et al
.
Urgency and necessity of Epstein-Barr virus prophylactic vaccines
.
NPJ Vaccin
.
2022
;
7
(
1
):
159
.
89.
Sokal
EM
,
Hoppenbrouwers
K
,
Vandermeulen
C
, et al
.
Recombinant gp350 vaccine for infectious mononucleosis: a phase 2, randomized, double-blind, placebo-controlled trial to evaluate the safety, immunogenicity, and efficacy of an Epstein-Barr virus vaccine in healthy young adults
.
J Infect Dis
.
2007
;
196
(
12
):
1749
-
1753
.
90.
Jean-Pierre
V
,
Lupo
J
,
Buisson
M
,
Morand
P
,
Germi
R
.
Main targets of interest for the development of a prophylactic or therapeutic Epstein-Barr virus vaccine
.
Front Microbiol
.
2021
;
12
:
701611
.
91.
Kanekiyo
M
,
Bu
W
,
Joyce
MG
, et al
.
Rational design of an Epstein-Barr virus vaccine targeting the receptor-binding site
.
Cell
.
2015
;
162
(
5
):
1090
-
1100
.
92.
Zhong
L
,
Zhao
Q
,
Zeng
MS
,
Zhang
X
.
Prophylactic vaccines against Epstein–Barr virus
.
The Lancet
.
2024
;
404
(
10455
):
845
.
93.
Cui
X
,
Cao
Z
,
Chen
Q
,
Arjunaraja
S
,
Snow
AL
,
Snapper
CM
.
Rabbits immunized with Epstein-Barr virus gH/gL or gB recombinant proteins elicit higher serum virus neutralizing activity than gp350
.
Vaccine
.
2016
;
34
(
34
):
4050
-
4055
.
94.
Ulger Toprak
N
,
Yagci
A
,
Gulluoglu
BM
, et al
.
A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer
.
Clin Microbiol Infect
.
2006
;
12
(
8
):
782
-
786
.
95.
Dejea
CM
,
Fathi
P
,
Craig
JM
, et al
.
Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria
.
Science (1979)
.
2018
;
359
(
6375
):
592
-
597
.
96.
Wu
S
,
Rhee
KJ
,
Albesiano
E
, et al
.
A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses
.
Nat Med
.
2009
;
15
(
9
):
1016
-
1022
.
97.
Wu
S
,
Lim
KC
,
Huang
J
,
Saidi
RF
,
Sears
CL
.
Bacteroides fragilis enterotoxin cleaves the zonula adherens protein, E-cadherin
.
Proc Natl Acad Sci U S A
.
1998
;
95
(
25
):
14979
-
14984
.
98.
Wu
S
,
Morin
PJ
,
Maouyo
D
,
Sears
CL
.
Bacteroides fragilis enterotoxin induces c-Myc expression and cellular proliferation
.
Gastroenterology
.
2003
;
124
(
2
):
392
-
400
.
99.
Obiso
RJ
,
Azghani
AO
,
Wilkins
TD
.
The Bacteroides fragilis toxin fragilysin disrupts the paracellular barrier of epithelial cells
.
Infect Immun
.
1997
;
65
(
4
):
1431
-
1439
.
100.
Riegler
M
,
Lotz
M
,
Sears
C
, et al
.
Bacteroides fragilis toxin 2 damages human colonic mucosa in vitro
.
Gut
.
1999
;
44
(
4
):
504
-
510
.
101.
Chung
L
,
Thiele Orberg
E
,
Geis
AL
, et al
.
Bacteroides fragilis toxin coordinates a pro-carcinogenic inflammatory cascade via targeting of colonic epithelial cells
.
Cell Host Microbe
.
2018
;
23
(
2
):
203
-
214.e5
.
102.
Franco
AA
,
Buckwold
SL
,
Shin
JW
,
Ascon
M
,
Sears
CL
.
Mutation of the zinc-binding metalloprotease motif affects Bacteroides fragilis toxin activity but does not affect propeptide processing
.
Infect Immun
.
2005
;
73
(
8
):
5273
-
5277
.
103.
Pleguezuelos-Manzano
C
,
Puschhof
J
,
Rosendahl Huber
A
, et al
.
Mutational signature in colorectal cancer caused by genotoxic pks + E. coli
.
Nature
.
2020
;
580
(
7802
):
269
-
273
.
104.
Buc
E
,
Dubois
D
,
Sauvanet
P
, et al
.
High prevalence of mucosa-associated E. coli producing cyclomodulin and genotoxin in colon cancer
.
PLoS One
.
2013
;
8
(
2
):
e56964
.
105.
Nougayrede
JP
,
Homburg
S
,
Taieb
F
, et al
.
Escherichia coli induces DNA double-strand breaks in eukaryotic cells
.
Science (1979)
.
2006
;
313
(
5788
).
106.
Bossuet-Greif
N
,
Vignard
J
,
Taieb
F
, et al
.
The colibactin genotoxin generates DNA interstrand cross-links in infected cells
.
mBio
.
2018
;
9
(
2
):
e02393-17
.
107.
Cuevas-Ramos
G
,
Petit
CR
,
Marcq
I
,
Boury
M
,
Oswald
E
,
Nougayrède
JP
.
Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells
.
Proc Natl Acad Sci U S A
.
2010
;
107
(
25
):
11537
-
11542
.
108.
Jans
M
,
Kolata
M
,
Blancke
G
, et al
.
Colibactin-driven colon cancer requires adhesin-mediated epithelial binding
.
Nature
.
2024
;
635
(
8038
):
472
-
480
.
109.
Langermann
S
,
Palaszynski
S
,
Barnhart
M
, et al
.
Prevention of mucosal Escherichia coli infection by FimH-adhesin–based systemic vaccination
.
Science
.
1979
;
1997
:
276
.
110.
Langermann
S
,
Möllby
R
,
Burlein
JE
, et al
.
Vaccination with FimH adhesin protects cynomolgus monkeys from colonization and infection by uropathogenic Escherichia coli
.
J Infect Dis
.
2000
;
181
(
2
):
774
-
778
.
111.
Lee-Six
H
,
Olafsson
S
,
Ellis
P
, et al
.
The landscape of somatic mutation in normal colorectal epithelial cells
.
Nature
.
2019
;
574
(
7779
):
532
-
537
.
112.
Van der Weken
H
,
Cox
E
,
Devriendt
B
.
Advances in oral subunit vaccine design
.
Vaccines (Basel)
.
2020
;
9
(
1
):
1
-
23
.
113.
Lavelle
EC
,
Ward
RW
.
Mucosal vaccines — fortifying the frontiers
.
Nat Rev Immunol
.
2022
;
22
(
4
):
236
-
250
.
114.
Clemens
JD
,
Sack
DA
,
Harris
JR
, et al
.
Field trial of oral cholera vaccines in Bangladesh: results from three-year follow-up
.
The Lancet
.
1990
;
335
(
8684
):
270
-
273
.
115.
Lockhart
PB
,
Brennan
MT
,
Sasser
HC
,
Fox
PC
,
Paster
BJ
,
Bahrani-Mougeot
FK
.
Bacteremia associated with toothbrushing and dental extraction
.
Circulation
.
2008
;
117
(
24
):
3118
-
3125
.
116.
Castellarin
M
,
Warren
RL
,
Freeman
JD
, et al
.
Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma
.
Genome Res
.
2012
;
22
(
2
):
299
-
306
.
117.
Kostic
AD
,
Gevers
D
,
Pedamallu
CS
, et al
.
Genomic analysis identifies association of Fusobacterium with colorectal carcinoma
.
Genome Res
.
2012
;
22
(
2
):
292
-
298
.
118.
Mitsuhashi
K
,
Nosho
K
,
Sukawa
Y
, et al
.
Association of Fusobacterium species in pancreatic cancer tissues with molecular features and prognosis
.
Oncotarget
.
2015
;
6
(
9
):
7209
-
7220
.
119.
Huang
S
,
Chen
J
,
Lian
L
, et al
.
Intratumoral levels and prognostic significance of Fusobacterium nucleatum in cervical carcinoma
.
Aging
.
2020
;
12
(
22
):
23337
-
23350
.
120.
Abed
J
,
Emgård
JEM
,
Zamir
G
, et al
.
Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed Gal-GalNAc
.
Cell Host Microbe
.
2016
;
20
(
2
):
215
-
225
.
121.
Zhang
L
,
Leng
XX
,
Qi
J
, et al
.
The adhesin RadD enhances Fusobacterium nucleatum tumour colonization and colorectal carcinogenesis
.
Nat Microbiol
.
2024
;
9
(
9
):
2292
-
2307
.
122.
Yang
Y
,
Weng
W
,
Peng
J
, et al
.
Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating Toll-like receptor 4 signaling to nuclear factor−κB, and up-regulating expression of MicroRNA-21
.
Gastroenterology
.
2017
;
152
(
4
):
851
-
866.e24
.
123.
Yu
TC
,
Guo
F
,
Yu
Y
, et al
.
Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy
.
Cell
.
2017
;
170
(
3
):
548
-
563.e16
.
124.
Jiang
SS
,
Xie Y Le
,
Xiao
XY
, et al
.
Fusobacterium nucleatum-derived succinic acid induces tumor resistance to immunotherapy in colorectal cancer
.
Cell Host Microbe
.
2023
;
31
(
5
):
781
-
797.e9
.
125.
Chen
L
,
Zhao
R
,
Shen
J
, et al
.
Antibacterial Fusobacterium nucleatum-mimicking nanomedicine to selectively eliminate tumor-colonized bacteria and enhance immunotherapy against colorectal cancer
.
Adv Mater
.
2023
;
35
(
45
):
e2306281
.
126.
Casasanta
MA
,
Yoo
CC
,
Udayasuryan
B
, et al
.
Fusobacterium nucleatum host cell binding and invasion induces IL-8 and CXCL1 secretion that drives colorectal cancer cell migration
.
Sci Signal
.
2020
;
13
(
641
):
eaba9157
.
127.
Serna
G
,
Ruiz-Pace
F
,
Hernando
J
, et al
.
Fusobacterium nucleatum persistence and risk of recurrence after preoperative treatment in locally advanced rectal cancer
.
Ann Oncol
.
2020
;
31
(
10
):
1366
-
1375
.
128.
Mima
K
,
Nishihara
R
,
Qian
ZR
, et al
.
Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis
.
Gut
.
2016
;
65
(
12
):
1973
-
1980
.
129.
Thurnheer
T
,
Guggenheim
B
,
Gruica
B
,
Gmür
R
.
Infinite serovar and ribotype heterogeneity among oral Fusobacterium nucleatum strains?
.
Anaerobe
.
1999
;
5
(
2
):
79
-
92
.
130.
Eaden
JA
,
Abrams
KR
,
Mayberry
JF
.
The risk of colorectal cancer in ulcerative colitis: a meta-analysis
.
Gut
.
2001
;
48
(
4
):
526
-
535
.
131.
Schiller
JT
,
Castellsagué
X
,
Villa
LL
,
Hildesheim
A
.
An update of prophylactic human papillomavirus L1 virus-like particle vaccine clinical trial results
.
Vaccine
.
2008
;
26
(
suppl 10
):
K53
-
K61
.
132.
Sears
CL
.
Enterotoxigenic Bacteroides fragilis: a rogue among symbiotes
.
Clin Microbiol Rev
.
2009
;
22
(
2
):
349
-
369
.
133.
Riedel
S
.
Edward Jenner and the history of smallpox and vaccination
.
Proc
.
2005
;
18
(
1
):
21
-
25
.
134.
Weiss
RA
,
Esparza
J
.
The prevention and eradication of smallpox: a commentary on Sloane (1755) ‘An account of inoculation.’
.
Philos Trans R Soc Lond B Biol Sci
.
2015
;
370
(
1666
):
20140378
.
135.
Meyer
H
,
Ehmann
R
,
Smith
GL
.
Smallpox in the post-eradication Era
.
Viruses
.
2020
;
12
(
2
):
138
.
136.
Boylston
A
.
The origins of inoculation
.
J R Soc Med
.
2012
;
105
(
7
):
309
-
313
.
137.
Smith
KA
.
Edward Jenner and the small pox vaccine
.
Front Immunol
.
2011
;
2
(
JUN
):
21
.