Guest guest Posted September 6, 1998 Report Share Posted September 6, 1998 Sorry about the error. You have to be registered with Medscape to read this article. Anyway here it is: The Search for Vaccines Against Helicobacter pylori P. Monath, MD, K. Lee, PhD, H. Ermak, PhD, Gwendolyn A. Myers, DVM, A. Weltzin, PhD, J. sca, PhD, D. , Jr., PhD, Gopalan Soman, PhD, Hitesh Bhagat, PhD, A. Ackerman, MD, Harry K. Kleanthous, PhD [infect Med 15(8):534-535,539-546, 1998. © 1998 SCP Communications, Inc.] Abstract Current antibiotic regimens against Helicobacter pylori are effective, but complex dosing and development of resistance are concerns. Animal studies and limited clinical trials of H pylori urease and other bacterial antigens have been conducted, with promising findings. [infect Med 15(8):534-535,539-546, 1998] Introduction One of the most promising recent developments in medicine is the concept that chronic afflictions, such as peptic ulcer disease and cancer can be controlled through immunization like classic infectious diseases. Research on vaccines against Helicobacter pylori-the leading cause of chronic gastritis and peptic ulcer disease and a primary risk factor for gastric adenocarcinoma-began in 1990. The favored approach has been the oral administration of purified recombinant subunit proteins of H pylori and a mucosal adjuvant, the labile toxin (LT) of Escherichia coli. As a single-component vaccine, the urease protein has shown remarkable prophylactic and therapeutic activity in animal models and partial therapeutic activity in humans. A number of other H pylori antigens have been effective in animal models, and the recent sequencing of the complete H pylori genome has led to an intensive effort in antigen discovery. Other research is directed at the comparison of adjuvants and vaccine delivery systems and toward the immunologic mechanisms mediating protection. Here, we present preclinical data, the results of early-stage clinical trials, and directions for future research on Helicobacter vaccines. Helicobacter pylori: Medical Impact A gram-negative spiral bacterium that specifically infects the stomach, H pylori (Fig. 1) is one of the most prevalent infections of humankind: Approximately 50% of adults in the industrialized world and more than 90% of inhabitants of developing countries are infected.[1] H pylori is thought to be acquired by person-to-person spread via the fecal-oral and oral-oral routes, and in some areas it may be waterborne.[2] After oral ingestion, the bacteria colonize gastric mucus in close association with gastric epithelial cells (Figs. 2,3). Infection is chronic and generally lifelong. In the US, approximately 2.5 million new H pylori infections occur each year. In Europe, the prevalence and incidence of H pylori-associated diseases are similar to or higher than those in the US. In industrialized countries, the incidence of infection is decreasing overall, although transmission varies with socioeconomic status, and subpopulations are thus differentially affected. H pylori is the cause of chronic gastritis and the vast majority of cases of peptic ulcer disease.[3-6] Conclusive evidence also exists for an etiologic role of H pylori infection in dysplasia and metaplasia of gastric mucosa, distal gastric adenocarcinoma, and non-Hodgkin's lymphoma of the stomach,[7-12] leading the World Health Organization to classify the bacteria as a Class I (definite) carcinogen.[13] Considered in terms of lifetime morbidity, the illness to infection ratio in the US and Europe may be estimated at 1:5 for peptic ulceration and 1:200 for gastric adenocarcinoma. Peptic Ulcer In the US, approximately 16,000 deaths are attributed annually to complications of peptic ulcer disease. There are more than 2 million physician visits per year for duodenal ulcers, 90% of which are attributable to H pylori, and more than 3 million physician visits per year for gastric ulcers, 60% of which are attributable to the bacterium.[1,14] In a prospective study, the risk of developing duodenal ulcer disease in H pylori-infected patients followed for 10 years exceeded 10%; in contrast, it was less than 1% in uninfected patients.[15] Gastric Adenocarcinoma The incidence of gastric adenocarcinoma in the US is approximately 24,000 cases per year, with 13,300 deaths, approximately 60% of which (14,400 cases; 7980 deaths) may be attributed to H pylori. The risk of developing gastric cancer is estimated to be 3- to 6-fold higher in infected than in uninfected individuals.[7,12,16] Gastric cancer is a leading cause of death in Latin America and Asia. Acquisition of H pylori infection early in life appears to be associated with early-onset gastric corpus atrophy and metaplasia and a higher risk of cancer.[17] Ingestion of dietary carcinogens and deficiencies in dietary antioxidants are thought to be important cofactors in the genesis of Helicobacter-related cancer. Helicobacter strain differences in virulence factors also appear to determine cancer risk.[18] Rationale for Vaccine Development Several lines of evidence provide a rationale for the development of a vaccine against H pylori. High Illness to Infection Ratio Compared with illness to infection ratios of other infectious diseases, that of H pylori-associated peptic ulcer is high (1:5). In comparison, such ratios are about 1:25 for hepatitis B-associated chronic liver disease and about 1:10 for Mycobacterium tuberculosis. Although the ratio for H pylori-associated gastric adenocarcinoma is lower (1:200), the morbidity and mortality associated with this disease are substantial. Vaccines for Prevention Vaccines have long been regarded as the most effective and economical approach to the prevention and control of infectious diseases. Although effective antimicrobial treatment for H pylori is now employed widely to prevent recurrence in patients with active or recent duodenal ulcer disease, the ability to treat does not obviate the need for preventive strategies. Indeed, most H pylori infections leading to gastric cancer and 20% to 30% of cases of upper gastrointestinal hemorrhage occur in individuals who have sustained long-term infections without antecedent symptoms. For this reason, these individuals do not present to the physician in time for antimicrobial intervention. Vaccines as Therapy or Complement Although the application of vaccines for therapy of infectious diseases is in its infancy, it has tremendous implications for the management of chronic infections, such as H pylori, HIV, human papillomavirus, viral hepatitis, herpesviruses, chlamydia, and a wide range of parasitic infections. From a practical perspective, the effectiveness of conventional antimicrobials has diminished interest in therapeutic vaccines. However, vaccines used in combination with antibiotics could improve the rate of treatment success and decrease the evolution of antimicrobial resistance and disease recurrence. Infection-induced immunity to H pylori is clearly insufficient to prevent reinfection, as shown by experiments in animals[19] and limited studies of humans.[20] In areas of the world with high rates of transmission of H pylori, re-infection may occur rapidly after treatment with antimicrobial agents. However, in industrialized nations, re-infection rates in adults appear to be low, both overall (0.5%-2%) and in high-risk individuals (2.5% in spouses of infected persons).[21] However, even in industrialized nations, re-infection rates in children may be substantially higher; in 1 study, Oderda and colleagues[22] reported that 18% of children became re-infected within 18 months of antibiotic therapy. Convenient Identification ofH pylori A wide array of simple office-based serologic screening tests and the noninvasive[13]C-urea breath test are now available for identifying infected individuals, and new serologic tests that identify H pylori strains characterized by a higher virulence phenotype, especially CagA, are under development.[23] These methods could be used to identify persons with H pylori gastritis during the first 2 decades of life, thus identifying a population at future risk of ulcer disease and cancer. If treatment of the infection is considered, coadministration of a vaccine to prevent re-infection will be an important component of such a strategy. Cost-effectiveness ofH pylori Vaccines Although there is convincing evidence for the cost-effectiveness of curing H pylori in patients with duodenal ulcer disease,[24] the pharmacoeconomics of prophylactic immunization-whether primary (pre-exposure) immunization or immunization to prevent re-infection-have not been well defined. Pre-exposure immunization requires application during infancy or childhood, depending on age of acquisition of infection in the population at risk. Since the indication for H pylori vaccination is the prevention of chronic diseases that occur in the third to the sixth decade of life, the cost-benefit ratio is influenced by heavy discounting of future cost savings from disease prevention. However, childhood immunization to prevent chronic disease acquired decades later is not without precedent and underlies the recommendation for universal immunization against hepatitis B,[25] a disease that causes considerably less cancer morbidity and mortality than H pylori.[26] In areas where gastric cancer is a leading cause of death, such as Latin America and Asia, individuals and society place a high value on investments that reduce the incidence of this incurable and fatal disease. The World Health Organization estimates that 550,000 gastric cancer deaths due to H pylori occur annually,[13] and these deaths must be considered potentially preventable through immunization. By way of comparison, 316,000 cases of hepatocellular carcinoma caused by hepatitis B occur annually, and many countries are implementing routine childhood immunization policies. Initial Immunization Trials Serious consideration of vaccination as a means to control peptic ulcer disease began around 1990. Pallen and Clayton[27] suggested that urease would be a candidate antigen for incorporation in an H pylori vaccine, based in part on findings in animals and humans immunized with jack-bean urease to suppress ammonia production in the intestine by ureolytic bacteria. Czinn and Nedrud[28] showed that H pylori whole-cell sonicates administered intragastrically to mice and ferrets elicited serum and intestinal immunoglobulin (Ig) G and IgA antibodies. Subsequent studies by Chen and coworkers[29,30] and Czinn and colleagues[31] demonstrated that mice orally immunized with Helicobacter sonicates or whole cells and cholera toxin (CT) adjuvant were protected against challenge with Helicobacter felis, a species capable of infecting murine gastric mucosa. In addition, passive protection against challenge was demonstrated by the oral administration of an IgA monoclonal antibody, suggesting that the principal mediator of protection after active immunization may be secretory IgA. The protective monoclonal antibody later was shown to be specific for Helicobacter urease.[32] In 1994, Michetti and others[33] demonstrated that mice orally immunized with recombinant H pylori urease were protected against challenge with H felis. Protective determinants were present on both subunits (UreA and UreB) of the recombinant multimeric urease molecule. The recombinant protein is similar to native urease in multimeric structure, molecular mass (550kDa), and nano-particulate morphology.[34] The UreB subunit truncated at the amino terminus,[35] and multimeric urease that had been aggregated or heat-denatured retained prophylactic activity (OraVax, unpublished data, 1997). These studies clearly demonstrated that urease is remarkable among bacterial proteins in its stability and immunogenicity. A large body of data has now been accumulated from several laboratories confirming that H pylori urease administered mucosally to a variety of animals confers protection against challenge.[34,36,37] While initial immunization studies utilized H felis as the challenge bacterium, the subsequent development of mouse models of H pylori infection led to the confirmation that urease protected against the human pathogen.[38-40] In 1994, Doidge and colleagues[41] reported that mice with subchronic H felis infection cleared or had reduced infection after oral immunization with H felis whole-cell sonicates. Urease administered orally to mice experimentally infected with H felis[42] or ferrets naturally infected with Helicobacter mustelae[43] was shown to have significant therapeutic activity. These studies indicated that the up regulation of immunity to specific H pylori antigens may result in clearance of chronic infection. The role of mucosal immunity in protection against H pylori in humans is also supported by a study of infants in West Africa, where infection usually occurs within the first year of life. Infants of mothers with high titers of anti-Helicobacter IgA in breast milk had a significant delay in acquisition of H pylori infection.[44] Subsequent studies indicate that the principal antigen recognized by breast milk IgA is urease (J. , MD, The Royal n Infirmary, Newcastle Upon Tyne, England, personal communication, 1996). Approaches to Vaccine Development Although the feasibility of prophylactic and therapeutic immunization was established by these initial studies, procedures for the large-scale production of a safe and effective product are needed (Table I). The use of whole bacterial cells or cellular extracts is problematic, and while recombinant subunit vaccines (especially urease) are attractive alternatives, the identification of a full complement of protective antigens to be included in a recombinant vaccine remains a considerable challenge. However, the greatest problem for vaccine developers is the selection of an effective method for presenting antigens to the host's immune system in such a way that protective or therapeutic immune responses are elicited in the gastric mucosa. Since the mechanisms by which H pylori evades immunity and the roles of T and B cells in effector responses are poorly understood, purely empirical approaches have been applied to screen antigens, adjuvants, and delivery systems. Approaches using live H pylori strains, live vectors, and subunit antigens have also been explored. LiveH pylori Vaccines Effective live, attenuated oral vaccines have been developed to protect against several enteric bacterial infections, including typhoid, cholera, and Shigella. However, this approach poses certain serious difficulties in the case of H pylori: Immunity resulting from infection with wild strains of H pylori does not result in clearance or provide protection against superinfection with other H pylori strains, recrudescence after antibiotic suppression, or re-infection after successful cure. A live, attenuated vaccine would probably elicit an even weaker immune response than the wild-type bacteria. Thus, it would be technically difficult to modify H pylori to induce effective immunity rather than the evasion or down-regulation of immunity associated with natural infection. It is likely that a live vaccine would require high doses (possibly >/=109 organisms) and repeated administrations to be effective. Therefore, high-yield fermentation of H pylori is difficult and may not be economically feasible at the scale required for a live vaccine. H pylori is well adapted to cause chronic, persistent infection in the host. Since human host responses are highly variable and uncontrollable, an attenuated vaccine must not cause persistent infection associated with an inflammatory response. Regulatory concerns about chronic infection with a vaccine strain would require long-term follow-up studies in large populations. The sensitivity of tests for persistence of a vaccine strain versus wild-type strains in humans is highly problematic. A live vaccine would elicit immune responses against a wide range of antigens, some of which may be undesirable, due to cross-reactivity with homologous human antigens or stimulation of delayed-type hypersensitivity responses. A live vaccine might be used as prophylaxis, but it is difficult to conceive of its use for treatment of infection. Despite these concerns, there may be a role for a live, attenuated H pylori vaccine in an effective prophylactic immunizing regimen. Preclinical studies in mice have demonstrated that H pylori-specific T and B cells are recruited to the gastric mucosa in large numbers only after Helicobacter challenge.[45] In mice immunized with urease before challenge, the gastric immune response is effective in clearing most of the challenge organisms, but without the stimulus provided by the challenge, the stomach remains immunologically silent. This observation suggests that an effective immunization might include priming of intestinal immunity with a subunit antigen, followed by a live, attenuated H pylori vaccine that would direct the immune response to the gastric mucosa but would establish only a transient infection sufficient to target immunity. The sequence of artificial immunizations in such a model may result in an immune response that is qualitatively distinct from natural infection. This concept is currently being explored in our laboratories. Live Vectors Recombinant enteric bacterial vectors have been constructed to deliver foreign antigens. Examples include attenuated strains of Shigella flexneri, Salmonella typhi, and E coli.[46] These vectors, as well as others that replicate in the gastrointestinal tract or invade the body by this route, provide potential approaches to immunization against mucosal pathogens such as H pylori. Examples of such vectors include Vibrio cholerae, Lactobacillus species, Streptococcus gordonii, poxvirus, adenovirus, poliovirus, rhinovirus, and alphavirus. The ideal live vector is one that is not replication-deficient or restricted in its ability to express its own and foreign antigenic determinants. Restriction of vector replication by anti-vector immunity is a concern that can potentially be addressed by a combination of 2 antigenically distinct vectors or a combination of parenteral priming followed by a live-vector boost or vice versa. Live vectors may preclude the need for a mucosal adjuvant by targeting M cells and inductive lymphoid tissues in the gut. Alternatively, the vectors may be designed to co-express antigens with immunomodulatory lymphokines. The use of live vectors could also simplify vaccine administration schedules, since fewer doses would be required than of a subunit vaccine. The manufacturing process is also greatly simplified, since protein purification is unnecessary. Preliminary studies have been performed in several laboratories with mixed results, and it is too early to draw conclusions about the value of live vectors for construction of an effective Helicobacter vaccine. Subunit Antigens Nonliving vaccines include defined subunits, whole-cell or crude preparations, and DNA-based vaccines. Whole-cell or crude preparations appear to be effective in animal models and have the advantage of multiple antigens presenting to the host without having to isolate, characterize, and prepare individually active components. This approach is unlikely to be practical from a scale-up perspective or desirable from a regulatory view, given the potential problem of autoimmunity due to Helicobacter antigens, such as blood group antigens (cross-reactive with human cells).[47,48] DNA-based approaches are being investigated, but it is too early to assess the feasibility of generating an effective mucosal (and especially gastric) immune response by this method. A nearer-term approach is the delivery of defined H pylori protein antigens in a formulation designed to elicit protective responses in the stomach. H pylori bacteria have a number of virulence factors that are of known importance in chronic infection, recruitment of inflammatory cells, and damage to mucosal epithelium (Fig. 1). Among these, prominent is the urease enzyme, which is implicated in acid tolerance of the bacteria, colonization, and mucin depletion. As noted, recombinant urease has been demonstrated to be highly effective in prophylactic immunization of mice against challenge with Helicobacter species.[33-41] Evidence of protection has also been obtained in models using larger animals, including cats and nonhuman primates.[49-51] Native urease is a metalloenzyme, dependent for enzymatic activity on Ni2+, incorporated during intracellular synthesis.[50] Urease is essential for colonization of the stomach by H pylori; the enzyme splits urea present in gastric juice to form ammonia, a strong base that presumably protects the bacterium from inactivation by gastric acid.[51,52] All strains of H pylori that infect humans express the urease enzyme. In fact, urease accounts for more than 6% of the total soluble bacterial protein of H pylori and is localized, in part, on the surface of the bacterium.[50,53,54] This makes the urease enzyme an important target for the immune response elicited by a vaccine. Urease is constitutively expressed in vivo so that the bacteria would be exposed to the anti-urease immune response during the entire course of infection. Moreover, H pylori urease is intrinsically acid-stable, making it an ideal vaccine for oral application. H pylori urease is highly conserved at the amino acid sequence level, and antigenic variation between strains of H pylori urease is not likely to impair vaccine efficacy. Cross-reactivity between the ureases of different H pylori clinical isolates and between H pylori urease and heterologous ureases of H felis and H mustelae has been demonstrated[52] and is the basis for the heterologous cross-protection studies.[31] In its native form, urease is a hexameric structure of large molecular mass (550kDa), composed of 6 copies of the UreA (30kDa) and UreB (60kDa) and has a particulate structure of 12nm in diameter,[49,50,55] favoring uptake by M cells in the gastrointestinal tract for induction of mucosal immunity.[56] The vaccine candidate-recombinant urease-is urease antigen produced in genetically engineered E coli. Antigenically indistinguishable from native urease, recombinant urease has an identical particulate structure but is enzymatically nonfunctional and does not generate toxic ammonia in the presence of urea. This has been accomplished by cloning and expressing in E coli only the genes for the structural subunits (ureA and ureB), omitting all other genes of the operon,[57] and including those involved in insertion of Ni2+ required for enzymatic function. After expression in fermentation cultures of E coli, the recombinant antigen is purified from bacterial lysates and is subsequently lyophilized in a stabilizer. Therapeutic Immunization Treatment of H pylori infection in patients with peptic ulcer disease is now an accepted health practice in the US[6] and Europe and is the basis for regulatory labeling of antibiotic-antisecretory drug combinations. However, antimicrobial therapy has a number of inherent limitations that might be overcome by use of an effective vaccine or a combined regimen of antibiotics and vaccine. On average, primary treatment failures occur in 15% of patients treated with antibiotics combined with an antisecretory drug. Poor compliance with complex antibiotic regimens and antibiotic resistance in H pylori[58-60] contribute to treatment failures. In contrast to antibiotics, vaccine-induced immunity is not expected to select for resistant or more virulent organisms. Since immunologic mechanisms are distinct from those involved in antimicrobial treatment, vaccines alone or synergistic activities of vaccines and antimicrobials could achieve the ultimate goal of 100% cure. Murine Studies Using recombinant urease[42,61] and crude cell antigens,[41] therapeutic activity has been documented in mice, with efficacy rates (determined by gastric urease activity) between 50% and 94%. When vaccine and a partially effective antibiotic regimen were combined, the latter proved to be more effective than either treatment alone.[62] These studies were conducted in mice with subchronic H felis infection, the immunization regimen being applied only a few weeks after infecting the animals. It is uncertain whether treatment would be as effective in a chronically infected host. Moreover, the reported cure rates based on gastric urease or histologic endpoints overestimate the effectiveness of immunization. In addition, in the mouse model, H felis is easier to eradicate than H pylori. The results with vaccine are also supported by the observation that mice can be cured of H felis with a single antibiotic,[63] whereas multiple drugs were required to achieve partial cure of H pylori.[64] When the H pylori mouse model was employed and therapeutic activity of urease-LT immunization was measured by quantitative culture, a statistically significant (P = 0.0016) 10-fold reduction in bacterial density (not eradication of infection) was observed. Interestingly, the LT adjuvant alone appeared to have some effect in reducing infection, possibly due to modulation of the immune response to antigens associated with natural infection. Ferret Studies In ferrets, immunization with urease and CT adjuvant resulted in presumptive cure of chronic H mustelae infection.[43] When tested 6 weeks after immunization, 30% of the ferrets were cured of infection. A significant reduction in gastric inflammation was demonstrated by histopathology in up to 60% of the animals. Interestingly, gastric inflammation was significantly reduced in the cured and persistently infected vaccinated animals compared with infected controls, a finding similar to that described in the rhesus monkeys.[72] The possibility that vaccines can diminish the pathologic consequences of Helicobacter infections deserves further study. Adjuvants All preclinical studies reported to date have demonstrated efficacy of vaccination against Helicobacter infection, using antigens given mucosally together with CT or LT as a mucosal adjuvant. No protection was achieved when antigens were administered without a mucosal adjuvant, even at exceedingly high levels.[34] CT is not acceptable as a human adjuvant because it induces diarrhea in humans at microgram levels.[65] LT is less reactogenic and has been tested clinically.[66] A possible means to circumvent the reactogenicity of native toxins as adjuvants is the use of atoxic cholera toxin B subunit (CTB) spiked with a low dose of native toxin. This combination was shown to be an effective adjuvant for an H felis sonicate vaccine, providing protection against H felis challenge.[34] An even more attractive approach is the use of genetically detoxified LT molecules, which are enzymatically inactive but still retain adjuvanticity.[67,68] Many novel adjuvants have shown promise in preclinical studies with a variety of other vaccines, including oil emulsions, saponins, immunostimulating complexes, polyphosphazine, muramyl dipeptide derivatives, block polymers, vitamin D3, liposomes, copolymer microspheres, and cytokines. Some data are now available from clinical trials; more is known about many of these adjuvants for parenteral than for mucosal routes of administration. In studies of H pylori urease antigen, a muramyl dipeptide derivative (N-acetylglucosaminyl-N-acetyl-muramyl-L-alanyl-D-isoglutamine, GMDP) delivered orally did not elicit protection in mice against challenge with H felis.[34] Alum given parenterally with urease was partially effective when given prophylactically (OraVax, unpublished data, 1997). An exploration of various adjuvants for parenteral immunization with urease and for combined mucosal-parenteral immunization regimens is currently underway in our laboratory and that of our partner, Pasteur Merieux Connaught. Preliminary data indicate that partial protection is achieved by parenteral injection of antigen with alum and other select adjuvants. Since adjuvants orient the immune response in a selective fashion with respect to T-helper subsets, the results of comparative studies will shed light on the role of Th1 and Th2 responses in protection. Immunization studies of interleukin-4 knock-out and gamma-interferon receptor deficient mice indicate that both Th1 and Th2 responses are required for protective immunity.[69] This finding is also supported by our observations of adjuvants having selective immunomodulatory properties. Clinical Trials Clinical testing of recombinant urease was initiated by our group in 1994, and trials of whole-cell and other recombinant antigens are in the planning stages by others. Our clinical studies were begun in healthy infected volunteers (rather than uninfected subjects) because of concern that immunization of naive individuals may potentiate inflammation upon subsequent infection. This phenomenon was at that time observed in mice[33,42,70] but subsequently not observed in cats or monkeys. In addition, because the immune correlates of protection remain problematic, it was believed that the direct measurement of a therapeutic effect in infected subjects would have the greatest clinical significance. A limited study was first performed to demonstrate the safety and tolerability of oral administration of urease without a mucosal adjuvant.[71] In a randomized, double-blind, placebo-controlled trial conducted by Kreiss and colleagues,[71] 6 infected asymptomatic adults were administered 4 doses of vaccine-each consisting of 60mg of recombinant H pylori urease-by the oral route once a week. Six infected subjects received placebo. As expected in the absence of an adjuvant, none of the vaccinated individuals mounted an immune response, and in gastric biopsies obtained before and 1 month after vaccination, no change in bacterial density (measured by quantitative culture), inflammation, or mucosal damage was observed. No adverse events was attributable to administration of urease. A second trial was conducted to determine the tolerability of coadministration of urease with a mucosal adjuvant (LT) in healthy adults with H pylori infection and to obtain preliminary data on therapeutic activity. Preliminary results of this trial-which was conducted at the Centre Hospitalier Universitaire, Lausanne, and at the Center for Vaccine Development, University of land in Baltimore-were reported by Michetti and colleagues at the Helicobacter congress in Copenhagen in 1996. Native LT purified from E coli was supplied by the Naval Medical Research Institute in Bethesda, land, which previously reported adjuvant activity in a study involving cholera vaccine.[66] The controlled trial involved administration of 4 weekly, graded doses of urease (20, 60, or 180mg) with LT; placebo vaccine with LT; or placebo vaccine and placebo adjuvant to groups of 4 or 5 volunteers. The ELISPOT assay for antibody-secreting cells (ASCs) in peripheral blood was the most sensitive determinant of immunologic response to the vaccine. Six of 14 (43%) subjects who received urease, but none of the 10 subjects who received placebo vaccine, had an increase in IgA or IgG ASCs at 1 or more time points, measured 7 days after each successive dose of vaccine. Gastric biopsies were obtained before and 1 month after completion of the immunization regimen. Differences were determined between pre- and postimmunization H pylori densities in gastric mucosa. Pairwise treatment group comparisons were performed at baseline and on the change from baseline to postimmunization. In addition, the significance of the mean change from baseline to postimmunization was assessed within each treatment group. While the urease-treated groups were not significantly different from control groups with respect to the change from baseline to postimmunization, the subjects receiving active urease experienced, on average, a larger decrease in bacterial densities from baseline to postimmunization (P = 0.032) than did subjects receiving placebo (P = 0.425). While the study had small sample sizes per group and was not powered to detect significant differences between treatment groups, it provided the first clinical evidence for a therapeutic activity of oral urease with LT adjuvant. The duration of the study was not sufficient to assess whether administration of the vaccine was associated with a decrease in inflammation, as has been observed in ferrets[43] and rhesus monkeys.[72] Conclusions and Future Research A convincing body of data now exists supporting the potential for successful immunization against H pylori. However, we are still at a preliminary stage in clinical development. The best immunogens, the best mode of presentation, the number of doses needed, optimal age at immunization, expected benefit, cost-effectiveness, and other factors involved in vaccine development require further study. The complex pathogenesis of this infection,[3,73] including the presence of antigens on H pylori shared with the host (a mechanism for immune evasion),[48] demands novel approaches to the development of a final vaccine formulation. The selection of defined and well-characterized recombinant subunit antigens appears to be the most viable approach, and the urease antigen has so far proved most potent in eliciting protective immunity. It is reasonable to assume that more than 1 protective component is needed in a vaccine, and a number of such antigens in addition to urease have now been discovered. The sequence analysis of the entire H pylori genome by Tomb and colleagues[74] will enhance antigen discovery efforts. In addition to antigen composition, a successful vaccine must be delivered to the host in a manner that elicits protective (therapeutic) immunity, particularly immunity expressed at the site of bacterial colonization (gastric mucosa). The most appropriate means to achieve this end has not yet been fully defined. Mucosal routes of immunization with a classic mucosal adjuvant (LT) have yielded the best results, but prophylactic (therapeutic) activity remains incomplete. Research is needed on the mechanisms of protective immunity induced by vaccines, on the protein-specific immune responses to natural infection, and on the functional role of T cells. Such studies may provide important data that lead to novel immunization methods, as well as surrogate tests for protection that are useful in vaccine trials. Additional discussion of animal models for the development of Helicobacter vaccine can be found on Medscape (www.medscape.com). Acknowledgments Original work described in this paper was funded in part by Pasteur Merieux Connaught (PMC) and by the National Institutes of Health. The authors are grateful to PMC scientists, particularly Drs. Pierre Meulien, Marie- Quentin-Millet, Farukh Rizvi, Bruno Guy, Ling Lissolo, and Veronique Mazarin (PMC, Marcy l'Etoile, France) for their scientific input about the work and its interpretation. Drs. Pierre Michetti, Christiana Kreiss, Irene Couthesy-Theulaz, and Andre Blum (Centre Hospitalier Universitaire, Lausanne, Switzerland); Kotloff and Genevieve Losonsky (Center for Vaccine Development, University of land, Baltimore, Md.); and (University of land Medical Center, Baltimore, Md.) conducted the clinical trials reviewed in this paper. Drs. Czinn and Nedrud (Case-Western Reserve University, Cleveland, Ohio); Fox (Massachusetts Institute of Technology, Cambridge, Mass.); Andre Dubois (Uniformed Services University of the Health Sciences, Bethesda, Md.); Soike (Tulane University, Covington, La.); and ph Hill, Christian Stadtlander, Hal Farris, and Gangemi (Clemson University, Clemson, S.C.) made significant contributions in many aspects of the testing of Helicobacter vaccine candidates in animal models. The authors are especially grateful for the excellent assistance of OraVax personnel, including ph Simon, Kochi, Tibbitts, Ingrassia, Gray, Kathleen Georgokopoulos, Amal Al-Gawari, , Rue Ding, and Bruce Ekstein. References 1. DN, Parsonnet J: Epidemiology and natural history of Helicobacter pylori infection, in Blaser MJ, PD, Ravdin J (eds): Infections of the Gastrointestinal Tract. New York, Raven Press, 1994. 2.Klein PD, Graham DY, Gaillour A, et al: Water source as a risk factor for Helicobacter pylori infection in Peruvian children. Lancet 337:1503-1506, 1991. 3.Blaser MJ: Helicobacter pylori and the pathogenesis of gastroduodenal inflammation. J Infect Dis 161:626-633, 1990. 4.Cover TL, Blaser MJ: Helicobacter pylori and gastroduodenal disease. Annu Rev Med 43:135-145, 1992. 5.Solnick JV, Tompkins LS: Helicobacter pylori and gastroduodenal disease: Pathogenesis and host-parasite interaction. Infect Agents Dis 1:294-309, 1993. 6.National Institutes of Health: Consensus development conference statement: Helicobacter pylori in peptic ulcer disease. JAMA 272:65-69, 1994. 7.Eurogast Study Group: An international association between Helicobacter pylori infection and gastric cancer. Lancet 341:1359-1362, 1993. 8.Forman D, Webb P: Geographic distribution and association with gastric cancer, in Northfield TC, Mendall M, Goggin PM (eds): Helicobacter pylori Infection. Dordrecht, Germany, Kluwer Academic Publishers, 1993, pp 11-20. 9.Correa P: Human gastric carcinogenesis: A multistep and multifactorial process. Cancer Res 52:6735-6740, 1992. 10.Wotherspoon AC, Doglioni C, Diss TC, et al: Regression of primary low-grade B-cell gastric lymphoma of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori. Lancet 342:575-577, 1993. 11.Parsonnet J, Hansen S, L, et al: Helicobacter pylori infection and gastric lymphoma. N Engl J Med 18:1267-1271, 1994. 12.Parsonnet J, Samloff IM, LM, et al: Helicobacter pylori, pepsinogen and risk for gastric adenocarcinoma. Cancer Epidemiol Biomarkers Prev 2:1-7, 1993. 13.World Health Organization: The State of World Health. Geneva, World Health Organization, 1996, pp 59-62. 14.Goodwin CS, Worsley BW: Helicobacter pylori: Biology and Clinical Practice. Boca Raton, CRC Press, 1993, p 465. 15.Sipponen P: Natural history of gastritis and the relationship to peptic ulcer disease. Digestion 5:70-75, 1990. 16. HM: The epidemiology of Helicobacter pylori infection and its relation to gastric cancer, in Goodwin CS, Worsley BW (eds): Helicobacter pylori: Biology and Clinical Practice. Boca Raton, CRC Press, 1993, pp 95-114. 17.Sipponen P, Seppala K: Gastric carcinoma: Failed adaptation to Helicobacter pylori. Scand J Gastroenterol 27(suppl 1):S33-S38, 1992. 18.Blaser MJ, - G, Kleanthous H, et al: Infection with Helicobacter pylori strains possessing cagA is associated with an increased risk of developing adenocarcinoma of the stomach. Cancer Res 55:1-5, 1995. 19.Lee CK, Soike K, Tibbitts T, et al: Urease immunization protects against re-infection by Helicobacter pylori in rhesus monkeys. Gut 39(2)(suppl):A45, 1996. 20.Gilman RH, Leon-Barua R, -Ramos A, et al: Rapid recurrence of Helicobacter pylori infection in Peruvian patients after eradication therapy. Proc Clin Infect Dis J 1997 (in press). 21.Marshall BJ: Epidemiology of H pylori in western countries, in Hunt RH, Tytgat GNJ (eds): Helicobacter pylori: Basic Mechanisms Top Clinical Cure. Dordrecht, Germany, Kluwer Academic Publishers, 1994, pp 75-84. 22.Oderda G, Vaira D, Ainley C: Eighteen month follow-up of H pylori positive children treated with amoxicillin and tinidazole. Dig Dis Sci 36:572-576, 1992. 23.Cover TL, Glupczynski Y, Lage AP: Serologic detection of infection with cagA+ Helicobacter pylori strains. J Clin Microbiol 33:1496-1500, 1995. 24.Deltenre M, Jonsson B: Pharmacoeconomics. Curr Opin Gastroenterol 12(suppl 1):S45-S49, 1996. 25.US Public Health Service: Vaccination and prophylaxis against hepatitis B in children. Am Fam Physician 50:979-984, 1994. 26.US Public Health Service: Important Information About Hepatitis B, Hepatitis B Vaccine, and Hepatitis B Immune Globulin. Washington, DC: US Depart Health Hum Services, May 1992. 27.Pallen MJ, Clayton CL: Vaccination against Helicobacter pylori urease. Lancet 336:186-187, 1990. 28.Czinn SJ, Nedrud JG: Oral immunization against Helicobacter pylori. Infect Immun 59:2359-2363, 1991. 29.Chen M, Lee A: Vaccination possibilities and probabilities, in Northfield TC, Mandell M, Goggin PM (eds): Helicobacter pylori Infection. Dordrecht, Germany, Kluwer Academic Publishers, 1993, pp 158-169. 30.Chen M, Lee A, Hazell S: Immunization against gastric Helicobacter infection in a mouse Helicobacter felis model. Lancet 339:1120-1121, 1992. 31.Czinn SJ, Cai A, Nedrud JG: Protection of germ-free mice from infection by Helicobacter felis after active oral or passive IgA immunization. Vaccine 11:637-642, 1993. 32.Blanchard TG, Czinn SJ, Maurer R, et al: Urease specific monoclonal antibodies prevent Helicobacter felis infection in mice. 63:1394-1399, 1995. 33.Michetti P, Couthesy-Theulaz I, Davin C, et al: Immunization of BALB/c mice against Helicobacter felis infection with Helicobacter pylori urease. Gastroenterology 107:1002-1011, 1994. 34.Lee CK, Weltzin RA, WD Jr., et al: Oral immunization with recombinant Helicobacter pylori urease induces secretory IgA antibodies and protects mice from challenge with Helicobacter felis. J Infect Dis 172:161-172, 1995. 35.Dore-Davin C, Michetti P, Saraga E, et al: A 37 kDa fragment of UreB is sufficient to confer protection against Helicobacter felis infection in mice. Gastroenterology 110(suppl):A97, 1996. 36.Pappo J, WD Jr., Kabok Z, et al: Effect of oral immunization with recombinant urease on murine Helicobacter felis gastritis. Infect Immun 63:1246-1252, 1995. 37.Ferrero RL, Thiberge JM, Huerre M, et al: Recombinant antigens prepared from the urease subunits of Helicobacter spp.: Evidence of protection in a mouse model of gastric infection. Infect Immun 62:4981-4989, 1994. 38.Marchetti M, Arico B, Burroni D, et al: Development of a mouse model of Helicobacter pylori infection that mimics human disease. Science 267:1655-1658, 1995. 39.Kleanthous H, Tibbits T, Bakios TJ, et al: In vivo selection of a highly adapted H pylori isolate and the development of an H pylori mouse model for studying vaccine efficacy. Gut 37(suppl):A94, 1995. 40.Kleanthous H, Myers G, Georgokopoulos K, et al: Effect of route of mucosal immunization with recombinant urease on gastric immune responses and protection against Helicobacter pylori infection. Gut 39(suppl 2):A75, 1996. 41.Doidge C, Gust I, Lee A, et al: Therapeutic immunization against Helicobacter infection. Lancet 343:914-915, 1994. 42.Couthesy-Theulaz I, Porta N, Glauser M, et al: Oral immunization with Helicobacter pylori urease B subunit as a treatment against Helicobacter infection in mice. Gastroenterology 109:115-121, 1995. 43.Cuenca R, Blanchard TG, Czinn SJ, et al: Therapeutic immunization against Helicobacter mustelae in naturally infected ferrets. Gastroenterology 110:1770-1775, 1996. 44. JE, Austin S, Dale A, et al: Protection by human milk IgA against Helicobacter pylori infection in infancy. Lancet 342:121, 1993. 45.Ermak TH, Kleanthous HK, Myers G, et al: Oral immunization of mice with recombinant Helicobacter pylori urease: Corpus gastritis after challenge with H felis is due to residual bacteria. Immunol Cell Biol 75 (suppl 1):A92, 1997. 46.Noriega FR, Losonsky G, Wang JY, et al: Further characterization of aroA virG Shigella flexneri a strain CVD1203 as a mucosal Shigella vaccine and a live-vector vaccine for delivering antigens of enterotoxigenic Escherichia coli. Infect Immun 64:23-27, 1996. 47.Negrini R, Savio A, Poiesi C, et al: Antigenic mimicry between Helicobacter pylori and gastric mucosa in the pathogenesis of body atrophic gastritis. Gastroenterology 111:655-665, 1996. 48.Appelmelk BJ, Simoons-Smit I, Negrini R, et al: Potential role of molecular mimicry between Helicobacter pylori lipopolysaccharide and host blood group antigens in autoimmunity. Infect Immun 64:2031-2040, 1996. 49.Mobley HLT, Foxall PA: H pylori urease, in Hunt R, Tytgat G (eds): Helicobacter pylori: Basic Mechanisms to Clinical Care. Dordrecht, Germany, Kluwer Academic Publishers, 1994, pp 41-58. 50.Mobley HLT, Island MD, Hausinger RP: Molecular biology of microbial ureases. Microbiol Rev 59:451-480, 1995. 51.Ferrero RL, Lee A: The importance of urease in acid protection for the gastric-colonizing bacteria Helicobacter pylori and H felis sp nov. Microb Ecol Health Dis 4:121-134, 1991. 52.Hawtin PR, Stacey AR, Newell DG: Investigation of the structure and localization of the urease of Helicobacter pylori using monoclonal antibodies. J Gen Microbiol 136:1995-2000, 1990. 53.Phadnis SH, Parlow MH, Levy M, et al: Surface localization of Helicobacter pylori urease and a heat shock protein homologue requires bacterial autolysis. Infect Immun 64:905-912, 1996. 54.Gootz TD, - G, Clancy J, et al: Immunological and molecular characterization of Helicobacter felis urease. Infect Immun 62:793-798, 1994. 55.Dunn BE, CP, - G, et al: Purification and characterization of urease from Helicobacter pylori. J Biol Chem 265:9464-9469, 1990. 56.Neutra MR, Kraehenbuhl J-P: Transepithelial transport of proteins by intestinal epithelial cells, in Audus KL, Raub TJ (eds): Biological Barriers to Protein Delivery. New York, Plenum Press, 1993, pp 107-129. 57.Labigne A, Cussac V, Courcoux P: Shuttle cloning and nucleotide sequence of Helicobacter pylori genes responsible for urease activity. J Bacteriol 173:1920-1931, 1991. 58.Noach LA, Langenberg WL, Bertola MA, et al: Impact of metronidazole resistance on the eradication of Helicobacter pylori. Scand J Infect Dis 26:321-327, 1994. 59.Megraud F: Helicobacter pylori resistance to antibiotics, in Hunt RH, Tytgat GNJ (eds): Helicobacter pylori: Basic Mechanisms to Clinical Cure. Dordrecht, Germany, Kluwer Academic Publishers, 1994, pp 570-583. 60.Parasakthi N, Goh KL: Primary and acquired resistance to clarithromycin among Helicobacter pylori strains in Malaysia. Am J Gastroenterol 90:519, 1995. 61.Kleanthous H, Ermak T, Pappo J, et al: Oral immunization with recombinant Helicobacter pylori urease apoenzyme in the treatment of Helicobacter infection. Gut 37(suppl):A94, 1995. 62.Kleanthous H, Tibbitts T, Bakios J, et al: Oral immunization with recombinant urease combined with antimicrobial therapy augment clearance of an H felis infection in mice. Gut 39(suppl 2):A75, 1996. 63.Hook-Nikanne J, Aho P, Karkkainen P, et al: The Helicobacter felis mouse model in assessing anti-Helicobacter therapies and gastric mucosal prostaglandin E2 levels. Scand J Gastroenterol 31:334-338, 1996. 64.Dubois A, Fiala N, Heman-Ackah LM, et al: Natural gastric infection with Helicobacter pylori in monkeys: A model for human infection with spiral bacteria. Gastroenterology 106:1405-1417, 1994. 65.Levine MM, Black RE, Clements ML, et al: Volunteer studies in development of vaccines against cholera and enterotoxigenic Escherichia coli, in Holme T, Holmgren J, Merson M, et al (eds): Acute Enteric Infection in Children: New Prospects for Treatment and Prevention. Amsterdam, Elsevier, 1981, pp 443-459. 66. DA, Baqar S, Oplinger M, et al: Safety and adjuvant activity of native and mutant E coli heat-labile toxins. From Progress on Modulation of the Immune Response to Vaccine Antigens, a symposium of the International Association of Biology Standards Task Force on Vaccines, World Health Organization, Bergen, Norway, June 18-21, 1996. 67.Dickinson BL, Clements JD: Dissociation of Escherichia coli heat-labile enterotoxin adjuvanticity from ADP-ribosyltransferase activity. Infect Immun 63:1617-1623, 1995. 68.Tommaso AD, Saletti G, Pizza M, et al: Induction of antigen-specific antibodies in vaginal secretions by using a nontoxic mutant of heat-labile enterotoxin as a mucosal adjuvant. Infect Immun 64:974-979, 1996. 69.Radcliff FJ, Ramsay AJ, Lee A: A mixed Th1/Th2 response may be necessary for effective immunity against Helicobacter. Immunol Cell Biol 75(suppl 1):A90, 1997. 70.Ermak TH, Ding R, Ekstein B, et al: Oral immunization of mice with recombinant Helicobacter pylori urease: Corpus gastritis after challenge with H felis is due to the presence of residual bacteria. Gastroenterology 113:1118-1128, 1997. 71.Kreiss C, Buclin T, Cosma M, et al: Safety of oral immunization with recombinant urease in patients with Helicobacter pylori infection. Lancet 347:1630-1631, 1996. 72.Dubois A, Lee CK, Fiala N, et al: Immunization against natural Helicobacter pylori infection in rhesus monkeys. Gastroenterology, 1998. In press. 73.Labigne A, de Reuse H: Determinants of Helicobacter pylori pathogenicity. Infect Agents Dis 5:191-202, 1996. 74.Tomb JF, White O, Kerlavage AR, et al: The complete genome sequence of the gastric pathogen Helicobacter pylori. Nature 388(6642):539-547, 1997. Quote Link to comment Share on other sites More sharing options...
Guest guest Posted September 6, 1998 Report Share Posted September 6, 1998 What really makes me sick is that they are testing efficicy and safety of the new vaccines on mice, ferrets, monkeys...and even innocent people in third world countries (West Africa in this case):::not considering that these people or animals have rights just as we. What makes us superior to justify risking the lives and health of people in other countries and animals in labratories? T Since the begining of time, there has always been some kind of disease to help stabalize population: the plague, yellow fever, malaria, diptheria. These diseases eventually subsided on their own (without vaccination) and another was created in its place. I think there is much truth in the theory of 'survival of the fittest' ...by trying to control the natural order of things, you will be confronted with more devestating effects. Polio, tetanus, malaria--many of these diseases seem to be a thing of the past; but now we have more serious diseases such as AIDS, luekemia and cancer at an all time HIGH.... I think Mendelsohn has made a significant point when stating that vaccines are a medical time bomb. Thanks for the article ....although I cannot understand much of the medical jargon. Lana Mama of Cody Ukiah At 09:37 AM 9/6/98 -0600, you wrote: >From: Mom2Q <Mom2Q@...> > > >Sorry about the error. You have to be registered with Medscape to read this article. Anyway here it is: > >The Search for Vaccines Against Helicobacter pylori > > P. Monath, MD, K. Lee, PhD, H. Ermak, PhD, Gwendolyn A. Myers, DVM, >A. Weltzin, PhD, J. sca, PhD, D. , Jr., PhD, Gopalan Soman, PhD, Hitesh Bhagat, >PhD, A. Ackerman, MD, Harry K. Kleanthous, PhD > >[infect Med 15(8):534-535,539-546, 1998. © 1998 SCP Communications, Inc.] > >Abstract > >Current antibiotic regimens against Helicobacter pylori are effective, but complex dosing and development of resistance are >concerns. Animal studies and limited clinical trials of H pylori urease and other bacterial antigens have been conducted, with >promising findings. [infect Med 15(8):534-535,539-546, 1998] > >Introduction > >One of the most promising recent developments in medicine is the concept that chronic afflictions, such as peptic ulcer >disease and cancer can be controlled through immunization like classic infectious diseases. Research on vaccines against >Helicobacter pylori-the leading cause of chronic gastritis and peptic ulcer disease and a primary risk factor for gastric >adenocarcinoma-began in 1990. The favored approach has been the oral administration of purified recombinant subunit >proteins of H pylori and a mucosal adjuvant, the labile toxin (LT) of Escherichia coli. As a single-component vaccine, the >urease protein has shown remarkable prophylactic and therapeutic activity in animal models and partial therapeutic activity in >humans. A number of other H pylori antigens have been effective in animal models, and the recent sequencing of the >complete H pylori genome has led to an intensive effort in antigen discovery. Other research is directed at the comparison of >adjuvants and vaccine delivery systems and toward the immunologic mechanisms mediating protection. Here, we present >preclinical data, the results of early-stage clinical trials, and directions for future research on Helicobacter vaccines. > >Helicobacter pylori: Medical Impact > >A gram-negative spiral bacterium that specifically infects the stomach, H pylori (Fig. 1) is one of the most prevalent >infections of humankind: Approximately 50% of adults in the industrialized world and more than 90% of inhabitants of >developing countries are infected.[1] H pylori is thought to be acquired by person-to-person spread via the fecal-oral and >oral-oral routes, and in some areas it may be waterborne.[2] After oral ingestion, the bacteria colonize gastric mucus in close >association with gastric epithelial cells (Figs. 2,3). Infection is chronic and generally lifelong. > >In the US, approximately 2.5 million new H pylori infections occur each year. In Europe, the prevalence and incidence of H >pylori-associated diseases are similar to or higher than those in the US. In industrialized countries, the incidence of infection >is decreasing overall, although transmission varies with socioeconomic status, and subpopulations are thus differentially >affected. > >H pylori is the cause of chronic gastritis and the vast majority of cases of peptic ulcer disease.[3-6] Conclusive evidence also >exists for an etiologic role of H pylori infection in dysplasia and metaplasia of gastric mucosa, distal gastric adenocarcinoma, >and non-Hodgkin's lymphoma of the stomach,[7-12] leading the World Health Organization to classify the bacteria as a Class I >(definite) carcinogen.[13] Considered in terms of lifetime morbidity, the illness to infection ratio in the US and Europe may be >estimated at 1:5 for peptic ulceration and 1:200 for gastric adenocarcinoma. > >Peptic Ulcer > >In the US, approximately 16,000 deaths are attributed annually to complications of peptic ulcer disease. There are more >than 2 million physician visits per year for duodenal ulcers, 90% of which are attributable to H pylori, and more than 3 >million physician visits per year for gastric ulcers, 60% of which are attributable to the bacterium.[1,14] In a prospective study, >the risk of developing duodenal ulcer disease in H pylori-infected patients followed for 10 years exceeded 10%; in contrast, >it was less than 1% in uninfected patients.[15] > >Gastric Adenocarcinoma > >The incidence of gastric adenocarcinoma in the US is approximately 24,000 cases per year, with 13,300 deaths, >approximately 60% of which (14,400 cases; 7980 deaths) may be attributed to H pylori. The risk of developing gastric >cancer is estimated to be 3- to 6-fold higher in infected than in uninfected individuals.[7,12,16] Gastric cancer is a leading cause >of death in Latin America and Asia. Acquisition of H pylori infection early in life appears to be associated with early-onset >gastric corpus atrophy and metaplasia and a higher risk of cancer.[17] Ingestion of dietary carcinogens and deficiencies in >dietary antioxidants are thought to be important cofactors in the genesis of Helicobacter-related cancer. Helicobacter strain >differences in virulence factors also appear to determine cancer risk.[18] > >Rationale for Vaccine Development > >Several lines of evidence provide a rationale for the development of a vaccine against H pylori. > >High Illness to Infection Ratio > >Compared with illness to infection ratios of other infectious diseases, that of H pylori-associated peptic ulcer is high (1:5). In >comparison, such ratios are about 1:25 for hepatitis B-associated chronic liver disease and about 1:10 for Mycobacterium >tuberculosis. Although the ratio for H pylori-associated gastric adenocarcinoma is lower (1:200), the morbidity and >mortality associated with this disease are substantial. > >Vaccines for Prevention > >Vaccines have long been regarded as the most effective and economical approach to the prevention and control of infectious >diseases. Although effective antimicrobial treatment for H pylori is now employed widely to prevent recurrence in patients >with active or recent duodenal ulcer disease, the ability to treat does not obviate the need for preventive strategies. Indeed, >most H pylori infections leading to gastric cancer and 20% to 30% of cases of upper gastrointestinal hemorrhage occur in >individuals who have sustained long-term infections without antecedent symptoms. For this reason, these individuals do not >present to the physician in time for antimicrobial intervention. > >Vaccines as Therapy or Complement > >Although the application of vaccines for therapy of infectious diseases is in its infancy, it has tremendous implications for the >management of chronic infections, such as H pylori, HIV, human papillomavirus, viral hepatitis, herpesviruses, chlamydia, >and a wide range of parasitic infections. > >>From a practical perspective, the effectiveness of conventional antimicrobials has diminished interest in therapeutic vaccines. >However, vaccines used in combination with antibiotics could improve the rate of treatment success and decrease the >evolution of antimicrobial resistance and disease recurrence. Infection-induced immunity to H pylori is clearly insufficient to >prevent reinfection, as shown by experiments in animals[19] and limited studies of humans.[20] In areas of the world with high >rates of transmission of H pylori, re-infection may occur rapidly after treatment with antimicrobial agents. However, in >industrialized nations, re-infection rates in adults appear to be low, both overall (0.5%-2%) and in high-risk individuals >(2.5% in spouses of infected persons).[21] However, even in industrialized nations, re-infection rates in children may be >substantially higher; in 1 study, Oderda and colleagues[22] reported that 18% of children became re-infected within 18 >months of antibiotic therapy. > >Convenient Identification ofH pylori > >A wide array of simple office-based serologic screening tests and the noninvasive[13]C-urea breath test are now available for >identifying infected individuals, and new serologic tests that identify H pylori strains characterized by a higher virulence >phenotype, especially CagA, are under development.[23] These methods could be used to identify persons with H pylori >gastritis during the first 2 decades of life, thus identifying a population at future risk of ulcer disease and cancer. If treatment >of the infection is considered, coadministration of a vaccine to prevent re-infection will be an important component of such a >strategy. > >Cost-effectiveness ofH pylori Vaccines > >Although there is convincing evidence for the cost-effectiveness of curing H pylori in patients with duodenal ulcer disease,[24] >the pharmacoeconomics of prophylactic immunization-whether primary (pre-exposure) immunization or immunization to >prevent re-infection-have not been well defined. Pre-exposure immunization requires application during infancy or >childhood, depending on age of acquisition of infection in the population at risk. Since the indication for H pylori vaccination >is the prevention of chronic diseases that occur in the third to the sixth decade of life, the cost-benefit ratio is influenced by >heavy discounting of future cost savings from disease prevention. However, childhood immunization to prevent chronic >disease acquired decades later is not without precedent and underlies the recommendation for universal immunization against >hepatitis B,[25] a disease that causes considerably less cancer morbidity and mortality than H pylori.[26] In areas where gastric >cancer is a leading cause of death, such as Latin America and Asia, individuals and society place a high value on investments >that reduce the incidence of this incurable and fatal disease. The World Health Organization estimates that 550,000 gastric >cancer deaths due to H pylori occur annually,[13] and these deaths must be considered potentially preventable through >immunization. By way of comparison, 316,000 cases of hepatocellular carcinoma caused by hepatitis B occur annually, and >many countries are implementing routine childhood immunization policies. > >Initial Immunization Trials > >Serious consideration of vaccination as a means to control peptic ulcer disease began around 1990. Pallen and Clayton[27] >suggested that urease would be a candidate antigen for incorporation in an H pylori vaccine, based in part on findings in >animals and humans immunized with jack-bean urease to suppress ammonia production in the intestine by ureolytic bacteria. >Czinn and Nedrud[28] showed that H pylori whole-cell sonicates administered intragastrically to mice and ferrets elicited >serum and intestinal immunoglobulin (Ig) G and IgA antibodies. Subsequent studies by Chen and coworkers[29,30] and Czinn >and colleagues[31] demonstrated that mice orally immunized with Helicobacter sonicates or whole cells and cholera toxin >(CT) adjuvant were protected against challenge with Helicobacter felis, a species capable of infecting murine gastric >mucosa. In addition, passive protection against challenge was demonstrated by the oral administration of an IgA monoclonal >antibody, suggesting that the principal mediator of protection after active immunization may be secretory IgA. The protective >monoclonal antibody later was shown to be specific for Helicobacter urease.[32] > >In 1994, Michetti and others[33] demonstrated that mice orally immunized with recombinant H pylori urease were protected >against challenge with H felis. Protective determinants were present on both subunits (UreA and UreB) of the recombinant >multimeric urease molecule. The recombinant protein is similar to native urease in multimeric structure, molecular mass >(550kDa), and nano-particulate morphology.[34] The UreB subunit truncated at the amino terminus,[35] and multimeric urease >that had been aggregated or heat-denatured retained prophylactic activity (OraVax, unpublished data, 1997). These studies >clearly demonstrated that urease is remarkable among bacterial proteins in its stability and immunogenicity. > >A large body of data has now been accumulated from several laboratories confirming that H pylori urease administered >mucosally to a variety of animals confers protection against challenge.[34,36,37] While initial immunization studies utilized H felis >as the challenge bacterium, the subsequent development of mouse models of H pylori infection led to the confirmation that >urease protected against the human pathogen.[38-40] > >In 1994, Doidge and colleagues[41] reported that mice with subchronic H felis infection cleared or had reduced infection >after oral immunization with H felis whole-cell sonicates. Urease administered orally to mice experimentally infected with H >felis[42] or ferrets naturally infected with Helicobacter mustelae[43] was shown to have significant therapeutic activity. These >studies indicated that the up regulation of immunity to specific H pylori antigens may result in clearance of chronic infection. >The role of mucosal immunity in protection against H pylori in humans is also supported by a study of infants in West Africa, >where infection usually occurs within the first year of life. Infants of mothers with high titers of anti-Helicobacter IgA in >breast milk had a significant delay in acquisition of H pylori infection.[44] Subsequent studies indicate that the principal antigen >recognized by breast milk IgA is urease (J. , MD, The Royal n Infirmary, Newcastle Upon Tyne, England, >personal communication, 1996). > >Approaches to Vaccine Development > >Although the feasibility of prophylactic and therapeutic immunization was established by these initial studies, procedures for >the large-scale production of a safe and effective product are needed (Table I). The use of whole bacterial cells or cellular >extracts is problematic, and while recombinant subunit vaccines (especially urease) are attractive alternatives, the >identification of a full complement of protective antigens to be included in a recombinant vaccine remains a considerable >challenge. However, the greatest problem for vaccine developers is the selection of an effective method for presenting >antigens to the host's immune system in such a way that protective or therapeutic immune responses are elicited in the gastric >mucosa. Since the mechanisms by which H pylori evades immunity and the roles of T and B cells in effector responses are >poorly understood, purely empirical approaches have been applied to screen antigens, adjuvants, and delivery systems. >Approaches using live H pylori strains, live vectors, and subunit antigens have also been explored. > >LiveH pylori Vaccines > >Effective live, attenuated oral vaccines have been developed to protect against several enteric bacterial infections, including >typhoid, cholera, and Shigella. However, this approach poses certain serious difficulties in the case of H pylori: > > Immunity resulting from infection with wild strains of H pylori does not result in clearance or provide protection > against superinfection with other H pylori strains, recrudescence after antibiotic suppression, or re-infection after > successful cure. A live, attenuated vaccine would probably elicit an even weaker immune response than the wild-type > bacteria. Thus, it would be technically difficult to modify H pylori to induce effective immunity rather than the evasion > or down-regulation of immunity associated with natural infection. > > It is likely that a live vaccine would require high doses (possibly >/=109 organisms) and repeated administrations to be > effective. Therefore, high-yield fermentation of H pylori is difficult and may not be economically feasible at the scale > required for a live vaccine. > > H pylori is well adapted to cause chronic, persistent infection in the host. Since human host responses are highly > variable and uncontrollable, an attenuated vaccine must not cause persistent infection associated with an inflammatory > response. Regulatory concerns about chronic infection with a vaccine strain would require long-term follow-up studies > in large populations. The sensitivity of tests for persistence of a vaccine strain versus wild-type strains in humans is > highly problematic. > > A live vaccine would elicit immune responses against a wide range of antigens, some of which may be undesirable, > due to cross-reactivity with homologous human antigens or stimulation of delayed-type hypersensitivity responses. > > A live vaccine might be used as prophylaxis, but it is difficult to conceive of its use for treatment of infection. > >Despite these concerns, there may be a role for a live, attenuated H pylori vaccine in an effective prophylactic immunizing >regimen. Preclinical studies in mice have demonstrated that H pylori-specific T and B cells are recruited to the gastric >mucosa in large numbers only after Helicobacter challenge.[45] In mice immunized with urease before challenge, the gastric >immune response is effective in clearing most of the challenge organisms, but without the stimulus provided by the challenge, >the stomach remains immunologically silent. This observation suggests that an effective immunization might include priming of >intestinal immunity with a subunit antigen, followed by a live, attenuated H pylori vaccine that would direct the immune >response to the gastric mucosa but would establish only a transient infection sufficient to target immunity. The sequence of >artificial immunizations in such a model may result in an immune response that is qualitatively distinct from natural infection. >This concept is currently being explored in our laboratories. > >Live Vectors > >Recombinant enteric bacterial vectors have been constructed to deliver foreign antigens. Examples include attenuated strains >of Shigella flexneri, Salmonella typhi, and E coli.[46] These vectors, as well as others that replicate in the gastrointestinal >tract or invade the body by this route, provide potential approaches to immunization against mucosal pathogens such as H >pylori. Examples of such vectors include Vibrio cholerae, Lactobacillus species, Streptococcus gordonii, poxvirus, >adenovirus, poliovirus, rhinovirus, and alphavirus. The ideal live vector is one that is not replication-deficient or restricted in >its ability to express its own and foreign antigenic determinants. Restriction of vector replication by anti-vector immunity is a >concern that can potentially be addressed by a combination of 2 antigenically distinct vectors or a combination of parenteral >priming followed by a live-vector boost or vice versa. Live vectors may preclude the need for a mucosal adjuvant by >targeting M cells and inductive lymphoid tissues in the gut. Alternatively, the vectors may be designed to co-express antigens >with immunomodulatory lymphokines. The use of live vectors could also simplify vaccine administration schedules, since >fewer doses would be required than of a subunit vaccine. The manufacturing process is also greatly simplified, since protein >purification is unnecessary. > >Preliminary studies have been performed in several laboratories with mixed results, and it is too early to draw conclusions >about the value of live vectors for construction of an effective Helicobacter vaccine. > >Subunit Antigens > >Nonliving vaccines include defined subunits, whole-cell or crude preparations, and DNA-based vaccines. Whole-cell or >crude preparations appear to be effective in animal models and have the advantage of multiple antigens presenting to the host >without having to isolate, characterize, and prepare individually active components. This approach is unlikely to be practical >from a scale-up perspective or desirable from a regulatory view, given the potential problem of autoimmunity due to >Helicobacter antigens, such as blood group antigens (cross-reactive with human cells).[47,48] DNA-based approaches >are being investigated, but it is too early to assess the feasibility of generating an effective mucosal (and especially gastric) >immune response by this method. > >A nearer-term approach is the delivery of defined H pylori protein antigens in a formulation designed to elicit protective >responses in the stomach. H pylori bacteria have a number of virulence factors that are of known importance in chronic >infection, recruitment of inflammatory cells, and damage to mucosal epithelium (Fig. 1). Among these, prominent is the >urease enzyme, which is implicated in acid tolerance of the bacteria, colonization, and mucin depletion. As noted, >recombinant urease has been demonstrated to be highly effective in prophylactic immunization of mice against challenge with >Helicobacter species.[33-41] Evidence of protection has also been obtained in models using larger animals, including cats and >nonhuman primates.[49-51] > >Native urease is a metalloenzyme, dependent for enzymatic activity on Ni2+, incorporated during intracellular synthesis.[50] >Urease is essential for colonization of the stomach by H pylori; the enzyme splits urea present in gastric juice to form >ammonia, a strong base that presumably protects the bacterium from inactivation by gastric acid.[51,52] All strains of H pylori >that infect humans express the urease enzyme. In fact, urease accounts for more than 6% of the total soluble bacterial protein >of H pylori and is localized, in part, on the surface of the bacterium.[50,53,54] This makes the urease enzyme an important >target for the immune response elicited by a vaccine. Urease is constitutively expressed in vivo so that the bacteria would be >exposed to the anti-urease immune response during the entire course of infection. Moreover, H pylori urease is intrinsically >acid-stable, making it an ideal vaccine for oral application. H pylori urease is highly conserved at the amino acid sequence >level, and antigenic variation between strains of H pylori urease is not likely to impair vaccine efficacy. Cross-reactivity >between the ureases of different H pylori clinical isolates and between H pylori urease and heterologous ureases of H felis >and H mustelae has been demonstrated[52] and is the basis for the heterologous cross-protection studies.[31] In its native >form, urease is a hexameric structure of large molecular mass (550kDa), composed of 6 copies of the UreA (30kDa) and >UreB (60kDa) and has a particulate structure of 12nm in diameter,[49,50,55] favoring uptake by M cells in the gastrointestinal >tract for induction of mucosal immunity.[56] > >The vaccine candidate-recombinant urease-is urease antigen produced in genetically engineered E coli. Antigenically >indistinguishable from native urease, recombinant urease has an identical particulate structure but is enzymatically >nonfunctional and does not generate toxic ammonia in the presence of urea. This has been accomplished by cloning and >expressing in E coli only the genes for the structural subunits (ureA and ureB), omitting all other genes of the operon,[57] and >including those involved in insertion of Ni2+ required for enzymatic function. After expression in fermentation cultures of E >coli, the recombinant antigen is purified from bacterial lysates and is subsequently lyophilized in a stabilizer. > >Therapeutic Immunization > >Treatment of H pylori infection in patients with peptic ulcer disease is now an accepted health practice in the US[6] and >Europe and is the basis for regulatory labeling of antibiotic-antisecretory drug combinations. However, antimicrobial therapy >has a number of inherent limitations that might be overcome by use of an effective vaccine or a combined regimen of >antibiotics and vaccine. On average, primary treatment failures occur in 15% of patients treated with antibiotics combined >with an antisecretory drug. Poor compliance with complex antibiotic regimens and antibiotic resistance in H pylori[58-60] >contribute to treatment failures. In contrast to antibiotics, vaccine-induced immunity is not expected to select for resistant or >more virulent organisms. Since immunologic mechanisms are distinct from those involved in antimicrobial treatment, vaccines >alone or synergistic activities of vaccines and antimicrobials could achieve the ultimate goal of 100% cure. > >Murine Studies > >Using recombinant urease[42,61] and crude cell antigens,[41] therapeutic activity has been documented in mice, with efficacy >rates (determined by gastric urease activity) between 50% and 94%. When vaccine and a partially effective antibiotic >regimen were combined, the latter proved to be more effective than either treatment alone.[62] These studies were conducted >in mice with subchronic H felis infection, the immunization regimen being applied only a few weeks after infecting the >animals. It is uncertain whether treatment would be as effective in a chronically infected host. Moreover, the reported cure >rates based on gastric urease or histologic endpoints overestimate the effectiveness of immunization. In addition, in the >mouse model, H felis is easier to eradicate than H pylori. The results with vaccine are also supported by the observation >that mice can be cured of H felis with a single antibiotic,[63] whereas multiple drugs were required to achieve partial cure of >H pylori.[64] When the H pylori mouse model was employed and therapeutic activity of urease-LT immunization was >measured by quantitative culture, a statistically significant (P = 0.0016) 10-fold reduction in bacterial density (not eradication >of infection) was observed. Interestingly, the LT adjuvant alone appeared to have some effect in reducing infection, possibly >due to modulation of the immune response to antigens associated with natural infection. > >Ferret Studies > >In ferrets, immunization with urease and CT adjuvant resulted in presumptive cure of chronic H mustelae infection.[43] When >tested 6 weeks after immunization, 30% of the ferrets were cured of infection. A significant reduction in gastric inflammation >was demonstrated by histopathology in up to 60% of the animals. Interestingly, gastric inflammation was significantly >reduced in the cured and persistently infected vaccinated animals compared with infected controls, a finding similar to that >described in the rhesus monkeys.[72] The possibility that vaccines can diminish the pathologic consequences of Helicobacter >infections deserves further study. > >Adjuvants > >All preclinical studies reported to date have demonstrated efficacy of vaccination against Helicobacter infection, using >antigens given mucosally together with CT or LT as a mucosal adjuvant. No protection was achieved when antigens were >administered without a mucosal adjuvant, even at exceedingly high levels.[34] CT is not acceptable as a human adjuvant >because it induces diarrhea in humans at microgram levels.[65] LT is less reactogenic and has been tested clinically.[66] A >possible means to circumvent the reactogenicity of native toxins as adjuvants is the use of atoxic cholera toxin B subunit >(CTB) spiked with a low dose of native toxin. This combination was shown to be an effective adjuvant for an H felis >sonicate vaccine, providing protection against H felis challenge.[34] An even more attractive approach is the use of genetically >detoxified LT molecules, which are enzymatically inactive but still retain adjuvanticity.[67,68] > >Many novel adjuvants have shown promise in preclinical studies with a variety of other vaccines, including oil emulsions, >saponins, immunostimulating complexes, polyphosphazine, muramyl dipeptide derivatives, block polymers, vitamin D3, >liposomes, copolymer microspheres, and cytokines. Some data are now available from clinical trials; more is known about >many of these adjuvants for parenteral than for mucosal routes of administration. In studies of H pylori urease antigen, a >muramyl dipeptide derivative (N-acetylglucosaminyl-N-acetyl-muramyl-L-alanyl-D-isoglutamine, GMDP) delivered orally >did not elicit protection in mice against challenge with H felis.[34] Alum given parenterally with urease was partially effective >when given prophylactically (OraVax, unpublished data, 1997). An exploration of various adjuvants for parenteral >immunization with urease and for combined mucosal-parenteral immunization regimens is currently underway in our >laboratory and that of our partner, Pasteur Merieux Connaught. Preliminary data indicate that partial protection is achieved >by parenteral injection of antigen with alum and other select adjuvants. Since adjuvants orient the immune response in a >selective fashion with respect to T-helper subsets, the results of comparative studies will shed light on the role of Th1 and >Th2 responses in protection. Immunization studies of interleukin-4 knock-out and gamma-interferon receptor deficient mice >indicate that both Th1 and Th2 responses are required for protective immunity.[69] This finding is also supported by our >observations of adjuvants having selective immunomodulatory properties. > >Clinical Trials > >Clinical testing of recombinant urease was initiated by our group in 1994, and trials of whole-cell and other recombinant >antigens are in the planning stages by others. Our clinical studies were begun in healthy infected volunteers (rather than >uninfected subjects) because of concern that immunization of naive individuals may potentiate inflammation upon subsequent >infection. This phenomenon was at that time observed in mice[33,42,70] but subsequently not observed in cats or monkeys. In >addition, because the immune correlates of protection remain problematic, it was believed that the direct measurement of a >therapeutic effect in infected subjects would have the greatest clinical significance. > >A limited study was first performed to demonstrate the safety and tolerability of oral administration of urease without a >mucosal adjuvant.[71] In a randomized, double-blind, placebo-controlled trial conducted by Kreiss and colleagues,[71] 6 >infected asymptomatic adults were administered 4 doses of vaccine-each consisting of 60mg of recombinant H pylori >urease-by the oral route once a week. Six infected subjects received placebo. As expected in the absence of an adjuvant, >none of the vaccinated individuals mounted an immune response, and in gastric biopsies obtained before and 1 month after >vaccination, no change in bacterial density (measured by quantitative culture), inflammation, or mucosal damage was >observed. No adverse events was attributable to administration of urease. > >A second trial was conducted to determine the tolerability of coadministration of urease with a mucosal adjuvant (LT) in >healthy adults with H pylori infection and to obtain preliminary data on therapeutic activity. Preliminary results of this >trial-which was conducted at the Centre Hospitalier Universitaire, Lausanne, and at the Center for Vaccine Development, >University of land in Baltimore-were reported by Michetti and colleagues at the Helicobacter congress in >Copenhagen in 1996. Native LT purified from E coli was supplied by the Naval Medical Research Institute in Bethesda, >land, which previously reported adjuvant activity in a study involving cholera vaccine.[66] > >The controlled trial involved administration of 4 weekly, graded doses of urease (20, 60, or 180mg) with LT; placebo >vaccine with LT; or placebo vaccine and placebo adjuvant to groups of 4 or 5 volunteers. The ELISPOT assay for >antibody-secreting cells (ASCs) in peripheral blood was the most sensitive determinant of immunologic response to the >vaccine. Six of 14 (43%) subjects who received urease, but none of the 10 subjects who received placebo vaccine, had an >increase in IgA or IgG ASCs at 1 or more time points, measured 7 days after each successive dose of vaccine. Gastric >biopsies were obtained before and 1 month after completion of the immunization regimen. Differences were determined >between pre- and postimmunization H pylori densities in gastric mucosa. Pairwise treatment group comparisons were >performed at baseline and on the change from baseline to postimmunization. In addition, the significance of the mean change >from baseline to postimmunization was assessed within each treatment group. While the urease-treated groups were not >significantly different from control groups with respect to the change from baseline to postimmunization, the subjects >receiving active urease experienced, on average, a larger decrease in bacterial densities from baseline to postimmunization >(P = 0.032) than did subjects receiving placebo (P = 0.425). While the study had small sample sizes per group and was not >powered to detect significant differences between treatment groups, it provided the first clinical evidence for a therapeutic >activity of oral urease with LT adjuvant. The duration of the study was not sufficient to assess whether administration of the >vaccine was associated with a decrease in inflammation, as has been observed in ferrets[43] and rhesus monkeys.[72] > >Conclusions and Future Research > >A convincing body of data now exists supporting the potential for successful immunization against H pylori. However, we >are still at a preliminary stage in clinical development. The best immunogens, the best mode of presentation, the number of >doses needed, optimal age at immunization, expected benefit, cost-effectiveness, and other factors involved in vaccine >development require further study. > >The complex pathogenesis of this infection,[3,73] including the presence of antigens on H pylori shared with the host (a >mechanism for immune evasion),[48] demands novel approaches to the development of a final vaccine formulation. The >selection of defined and well-characterized recombinant subunit antigens appears to be the most viable approach, and the >urease antigen has so far proved most potent in eliciting protective immunity. It is reasonable to assume that more than 1 >protective component is needed in a vaccine, and a number of such antigens in addition to urease have now been >discovered. The sequence analysis of the entire H pylori genome by Tomb and colleagues[74] will enhance antigen discovery >efforts. In addition to antigen composition, a successful vaccine must be delivered to the host in a manner that elicits >protective (therapeutic) immunity, particularly immunity expressed at the site of bacterial colonization (gastric mucosa). The >most appropriate means to achieve this end has not yet been fully defined. Mucosal routes of immunization with a classic >mucosal adjuvant (LT) have yielded the best results, but prophylactic (therapeutic) activity remains incomplete. Research is >needed on the mechanisms of protective immunity induced by vaccines, on the protein-specific immune responses to natural >infection, and on the functional role of T cells. Such studies may provide important data that lead to novel immunization >methods, as well as surrogate tests for protection that are useful in vaccine trials. > >Additional discussion of animal models for the development of Helicobacter vaccine can be found on Medscape >(www.medscape.com). > >Acknowledgments > >Original work described in this paper was funded in part by Pasteur Merieux Connaught (PMC) and by the National >Institutes of Health. The authors are grateful to PMC scientists, particularly Drs. Pierre Meulien, Marie- Quentin-Millet, >Farukh Rizvi, Bruno Guy, Ling Lissolo, and Veronique Mazarin (PMC, Marcy l'Etoile, France) for their scientific input >about the work and its interpretation. Drs. Pierre Michetti, Christiana Kreiss, Irene Couthesy-Theulaz, and Andre Blum >(Centre Hospitalier Universitaire, Lausanne, Switzerland); Kotloff and Genevieve Losonsky (Center for Vaccine >Development, University of land, Baltimore, Md.); and (University of land Medical Center, >Baltimore, Md.) conducted the clinical trials reviewed in this paper. Drs. Czinn and Nedrud (Case-Western >Reserve University, Cleveland, Ohio); Fox (Massachusetts Institute of Technology, Cambridge, Mass.); Andre >Dubois (Uniformed Services University of the Health Sciences, Bethesda, Md.); Soike (Tulane University, >Covington, La.); and ph Hill, Christian Stadtlander, Hal Farris, and Gangemi (Clemson University, Clemson, >S.C.) made significant contributions in many aspects of the testing of Helicobacter vaccine candidates in animal models. The >authors are especially grateful for the excellent assistance of OraVax personnel, including ph Simon, Kochi, > Tibbitts, Ingrassia, Gray, Kathleen Georgokopoulos, Amal Al-Gawari, , Rue Ding, >and Bruce Ekstein. > >References > > 1. DN, Parsonnet J: Epidemiology and natural history of Helicobacter pylori infection, in Blaser MJ, PD, > Ravdin J (eds): Infections of the Gastrointestinal Tract. New York, Raven Press, 1994. > > 2.Klein PD, Graham DY, Gaillour A, et al: Water source as a risk factor for Helicobacter pylori infection in Peruvian > children. Lancet 337:1503-1506, 1991. > > 3.Blaser MJ: Helicobacter pylori and the pathogenesis of gastroduodenal inflammation. J Infect Dis 161:626-633, > 1990. > > 4.Cover TL, Blaser MJ: Helicobacter pylori and gastroduodenal disease. Annu Rev Med 43:135-145, 1992. > > 5.Solnick JV, Tompkins LS: Helicobacter pylori and gastroduodenal disease: Pathogenesis and host-parasite > interaction. Infect Agents Dis 1:294-309, 1993. > > 6.National Institutes of Health: Consensus development conference statement: Helicobacter pylori in peptic ulcer > disease. JAMA 272:65-69, 1994. > > 7.Eurogast Study Group: An international association between Helicobacter pylori infection and gastric cancer. Lancet > 341:1359-1362, 1993. > > 8.Forman D, Webb P: Geographic distribution and association with gastric cancer, in Northfield TC, Mendall M, > Goggin PM (eds): Helicobacter pylori Infection. Dordrecht, Germany, Kluwer Academic Publishers, 1993, pp > 11-20. > > 9.Correa P: Human gastric carcinogenesis: A multistep and multifactorial process. Cancer Res 52:6735-6740, 1992. > > 10.Wotherspoon AC, Doglioni C, Diss TC, et al: Regression of primary low-grade B-cell gastric lymphoma of > mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori. Lancet 342:575-577, 1993. > > 11.Parsonnet J, Hansen S, L, et al: Helicobacter pylori infection and gastric lymphoma. N Engl J Med > 18:1267-1271, 1994. > > 12.Parsonnet J, Samloff IM, LM, et al: Helicobacter pylori, pepsinogen and risk for gastric adenocarcinoma. > Cancer Epidemiol Biomarkers Prev 2:1-7, 1993. > > 13.World Health Organization: The State of World Health. Geneva, World Health Organization, 1996, pp 59-62. > > 14.Goodwin CS, Worsley BW: Helicobacter pylori: Biology and Clinical Practice. Boca Raton, CRC Press, 1993, p > 465. > > 15.Sipponen P: Natural history of gastritis and the relationship to peptic ulcer disease. Digestion 5:70-75, 1990. > > 16. HM: The epidemiology of Helicobacter pylori infection and its relation to gastric cancer, in Goodwin CS, > Worsley BW (eds): Helicobacter pylori: Biology and Clinical Practice. Boca Raton, CRC Press, 1993, pp 95-114. > > 17.Sipponen P, Seppala K: Gastric carcinoma: Failed adaptation to Helicobacter pylori. Scand J Gastroenterol > 27(suppl 1):S33-S38, 1992. > > 18.Blaser MJ, - G, Kleanthous H, et al: Infection with Helicobacter pylori strains possessing cagA is > associated with an increased risk of developing adenocarcinoma of the stomach. Cancer Res 55:1-5, 1995. > > 19.Lee CK, Soike K, Tibbitts T, et al: Urease immunization protects against re-infection by Helicobacter pylori in > rhesus monkeys. Gut 39(2)(suppl):A45, 1996. > > 20.Gilman RH, Leon-Barua R, -Ramos A, et al: Rapid recurrence of Helicobacter pylori infection in Peruvian > patients after eradication therapy. Proc Clin Infect Dis J 1997 (in press). > > 21.Marshall BJ: Epidemiology of H pylori in western countries, in Hunt RH, Tytgat GNJ (eds): Helicobacter pylori: > Basic Mechanisms Top Clinical Cure. Dordrecht, Germany, Kluwer Academic Publishers, 1994, pp 75-84. > > 22.Oderda G, Vaira D, Ainley C: Eighteen month follow-up of H pylori positive children treated with amoxicillin and > tinidazole. Dig Dis Sci 36:572-576, 1992. > > 23.Cover TL, Glupczynski Y, Lage AP: Serologic detection of infection with cagA+ Helicobacter pylori strains. J Clin > Microbiol 33:1496-1500, 1995. > > 24.Deltenre M, Jonsson B: Pharmacoeconomics. Curr Opin Gastroenterol 12(suppl 1):S45-S49, 1996. > > 25.US Public Health Service: Vaccination and prophylaxis against hepatitis B in children. Am Fam Physician > 50:979-984, 1994. > > 26.US Public Health Service: Important Information About Hepatitis B, Hepatitis B Vaccine, and Hepatitis B Immune > Globulin. Washington, DC: US Depart Health Hum Services, May 1992. > > 27.Pallen MJ, Clayton CL: Vaccination against Helicobacter pylori urease. Lancet 336:186-187, 1990. > > 28.Czinn SJ, Nedrud JG: Oral immunization against Helicobacter pylori. Infect Immun 59:2359-2363, 1991. > > 29.Chen M, Lee A: Vaccination possibilities and probabilities, in Northfield TC, Mandell M, Goggin PM (eds): > Helicobacter pylori Infection. Dordrecht, Germany, Kluwer Academic Publishers, 1993, pp 158-169. > > 30.Chen M, Lee A, Hazell S: Immunization against gastric Helicobacter infection in a mouse Helicobacter felis model. > Lancet 339:1120-1121, 1992. > > 31.Czinn SJ, Cai A, Nedrud JG: Protection of germ-free mice from infection by Helicobacter felis after active oral or > passive IgA immunization. Vaccine 11:637-642, 1993. > > 32.Blanchard TG, Czinn SJ, Maurer R, et al: Urease specific monoclonal antibodies prevent Helicobacter felis infection > in mice. 63:1394-1399, 1995. > > 33.Michetti P, Couthesy-Theulaz I, Davin C, et al: Immunization of BALB/c mice against Helicobacter felis infection > with Helicobacter pylori urease. Gastroenterology 107:1002-1011, 1994. > > 34.Lee CK, Weltzin RA, WD Jr., et al: Oral immunization with recombinant Helicobacter pylori urease > induces secretory IgA antibodies and protects mice from challenge with Helicobacter felis. J Infect Dis > 172:161-172, 1995. > > 35.Dore-Davin C, Michetti P, Saraga E, et al: A 37 kDa fragment of UreB is sufficient to confer protection against > Helicobacter felis infection in mice. Gastroenterology 110(suppl):A97, 1996. > > 36.Pappo J, WD Jr., Kabok Z, et al: Effect of oral immunization with recombinant urease on murine > Helicobacter felis gastritis. Infect Immun 63:1246-1252, 1995. > > 37.Ferrero RL, Thiberge JM, Huerre M, et al: Recombinant antigens prepared from the urease subunits of Helicobacter > spp.: Evidence of protection in a mouse model of gastric infection. Infect Immun 62:4981-4989, 1994. > > 38.Marchetti M, Arico B, Burroni D, et al: Development of a mouse model of Helicobacter pylori infection that mimics > human disease. Science 267:1655-1658, 1995. > > 39.Kleanthous H, Tibbits T, Bakios TJ, et al: In vivo selection of a highly adapted H pylori isolate and the development > of an H pylori mouse model for studying vaccine efficacy. Gut 37(suppl):A94, 1995. > > 40.Kleanthous H, Myers G, Georgokopoulos K, et al: Effect of route of mucosal immunization with recombinant urease > on gastric immune responses and protection against Helicobacter pylori infection. Gut 39(suppl 2):A75, 1996. > > 41.Doidge C, Gust I, Lee A, et al: Therapeutic immunization against Helicobacter infection. Lancet 343:914-915, 1994. > > 42.Couthesy-Theulaz I, Porta N, Glauser M, et al: Oral immunization with Helicobacter pylori urease B subunit as a > treatment against Helicobacter infection in mice. Gastroenterology 109:115-121, 1995. > > 43.Cuenca R, Blanchard TG, Czinn SJ, et al: Therapeutic immunization against Helicobacter mustelae in naturally > infected ferrets. Gastroenterology 110:1770-1775, 1996. > > 44. JE, Austin S, Dale A, et al: Protection by human milk IgA against Helicobacter pylori infection in infancy. > Lancet 342:121, 1993. > > 45.Ermak TH, Kleanthous HK, Myers G, et al: Oral immunization of mice with recombinant Helicobacter pylori urease: > Corpus gastritis after challenge with H felis is due to residual bacteria. Immunol Cell Biol 75 (suppl 1):A92, 1997. > > 46.Noriega FR, Losonsky G, Wang JY, et al: Further characterization of aroA virG Shigella flexneri a strain > CVD1203 as a mucosal Shigella vaccine and a live-vector vaccine for delivering antigens of enterotoxigenic > Escherichia coli. Infect Immun 64:23-27, 1996. > > 47.Negrini R, Savio A, Poiesi C, et al: Antigenic mimicry between Helicobacter pylori and gastric mucosa in the > pathogenesis of body atrophic gastritis. Gastroenterology 111:655-665, 1996. > > 48.Appelmelk BJ, Simoons-Smit I, Negrini R, et al: Potential role of molecular mimicry between Helicobacter pylori > lipopolysaccharide and host blood group antigens in autoimmunity. Infect Immun 64:2031-2040, 1996. > > 49.Mobley HLT, Foxall PA: H pylori urease, in Hunt R, Tytgat G (eds): Helicobacter pylori: Basic Mechanisms to > Clinical Care. Dordrecht, Germany, Kluwer Academic Publishers, 1994, pp 41-58. > > 50.Mobley HLT, Island MD, Hausinger RP: Molecular biology of microbial ureases. Microbiol Rev 59:451-480, 1995. > > 51.Ferrero RL, Lee A: The importance of urease in acid protection for the gastric-colonizing bacteria Helicobacter > pylori and H felis sp nov. Microb Ecol Health Dis 4:121-134, 1991. > > 52.Hawtin PR, Stacey AR, Newell DG: Investigation of the structure and localization of the urease of Helicobacter > pylori using monoclonal antibodies. J Gen Microbiol 136:1995-2000, 1990. > > 53.Phadnis SH, Parlow MH, Levy M, et al: Surface localization of Helicobacter pylori urease and a heat shock protein > homologue requires bacterial autolysis. Infect Immun 64:905-912, 1996. > > 54.Gootz TD, - G, Clancy J, et al: Immunological and molecular characterization of Helicobacter felis > urease. Infect Immun 62:793-798, 1994. > > 55.Dunn BE, CP, - G, et al: Purification and characterization of urease from Helicobacter pylori. J > Biol Chem 265:9464-9469, 1990. > > 56.Neutra MR, Kraehenbuhl J-P: Transepithelial transport of proteins by intestinal epithelial cells, in Audus KL, Raub TJ > (eds): Biological Barriers to Protein Delivery. New York, Plenum Press, 1993, pp 107-129. > > 57.Labigne A, Cussac V, Courcoux P: Shuttle cloning and nucleotide sequence of Helicobacter pylori genes > responsible for urease activity. J Bacteriol 173:1920-1931, 1991. > > 58.Noach LA, Langenberg WL, Bertola MA, et al: Impact of metronidazole resistance on the eradication of > Helicobacter pylori. Scand J Infect Dis 26:321-327, 1994. > > 59.Megraud F: Helicobacter pylori resistance to antibiotics, in Hunt RH, Tytgat GNJ (eds): Helicobacter pylori: Basic > Mechanisms to Clinical Cure. Dordrecht, Germany, Kluwer Academic Publishers, 1994, pp 570-583. > > 60.Parasakthi N, Goh KL: Primary and acquired resistance to clarithromycin among Helicobacter pylori strains in > Malaysia. Am J Gastroenterol 90:519, 1995. > > 61.Kleanthous H, Ermak T, Pappo J, et al: Oral immunization with recombinant Helicobacter pylori urease apoenzyme > in the treatment of Helicobacter infection. Gut 37(suppl):A94, 1995. > > 62.Kleanthous H, Tibbitts T, Bakios J, et al: Oral immunization with recombinant urease combined with antimicrobial > therapy augment clearance of an H felis infection in mice. Gut 39(suppl 2):A75, 1996. > > 63.Hook-Nikanne J, Aho P, Karkkainen P, et al: The Helicobacter felis mouse model in assessing anti-Helicobacter > therapies and gastric mucosal prostaglandin E2 levels. Scand J Gastroenterol 31:334-338, 1996. > > 64.Dubois A, Fiala N, Heman-Ackah LM, et al: Natural gastric infection with Helicobacter pylori in monkeys: A model > for human infection with spiral bacteria. Gastroenterology 106:1405-1417, 1994. > > 65.Levine MM, Black RE, Clements ML, et al: Volunteer studies in development of vaccines against cholera and > enterotoxigenic Escherichia coli, in Holme T, Holmgren J, Merson M, et al (eds): Acute Enteric Infection in > Children: New Prospects for Treatment and Prevention. Amsterdam, Elsevier, 1981, pp 443-459. > > 66. DA, Baqar S, Oplinger M, et al: Safety and adjuvant activity of native and mutant E coli heat-labile toxins. > From Progress on Modulation of the Immune Response to Vaccine Antigens, a symposium of the International > Association of Biology Standards Task Force on Vaccines, World Health Organization, Bergen, Norway, June > 18-21, 1996. > > 67.Dickinson BL, Clements JD: Dissociation of Escherichia coli heat-labile enterotoxin adjuvanticity from > ADP-ribosyltransferase activity. Infect Immun 63:1617-1623, 1995. > > 68.Tommaso AD, Saletti G, Pizza M, et al: Induction of antigen-specific antibodies in vaginal secretions by using a > nontoxic mutant of heat-labile enterotoxin as a mucosal adjuvant. Infect Immun 64:974-979, 1996. > > 69.Radcliff FJ, Ramsay AJ, Lee A: A mixed Th1/Th2 response may be necessary for effective immunity against > Helicobacter. Immunol Cell Biol 75(suppl 1):A90, 1997. > > 70.Ermak TH, Ding R, Ekstein B, et al: Oral immunization of mice with recombinant Helicobacter pylori urease: Corpus > gastritis after challenge with H felis is due to the presence of residual bacteria. Gastroenterology 113:1118-1128, > 1997. > > 71.Kreiss C, Buclin T, Cosma M, et al: Safety of oral immunization with recombinant urease in patients with > Helicobacter pylori infection. Lancet 347:1630-1631, 1996. > > 72.Dubois A, Lee CK, Fiala N, et al: Immunization against natural Helicobacter pylori infection in rhesus monkeys. > Gastroenterology, 1998. In press. > > 73.Labigne A, de Reuse H: Determinants of Helicobacter pylori pathogenicity. Infect Agents Dis 5:191-202, 1996. > > 74.Tomb JF, White O, Kerlavage AR, et al: The complete genome sequence of the gastric pathogen Helicobacter > pylori. Nature 388(6642):539-547, 1997. > > > > >------------------------------------------------------------------------ > Quote Link to comment Share on other sites More sharing options...
Guest guest Posted September 7, 1998 Report Share Posted September 7, 1998 In the same vein, check this book out: " The Fourth Horseman: A Short History of Plagues and Diseases " by Nikiforuk. I may not have the title exactly right. It's a fascinating and horrifying read, and talks about the devastating consequences of messing with the natural order of things. Kate At 01:42 AM 9/7/98 -0400, Mark & Lana Clifton wrote: >From: Mark & Lana Clifton <mclifton@...> > >What really makes me sick is that they are testing efficicy and safety of >the new vaccines on mice, ferrets, monkeys...and even innocent people in >third world countries (West Africa in this case):::not considering that >these people or animals have rights just as we. What makes us superior to >justify risking the lives and health of people in other countries and >animals in labratories? T > >Since the begining of time, there has always been some kind of disease to >help stabalize population: the plague, yellow fever, malaria, diptheria. >These diseases eventually subsided on their own (without vaccination) and >another was created in its place. I think there is much truth in the >theory of 'survival of the fittest' ...by trying to control the natural >order of things, you will be confronted with more devestating effects. >Polio, tetanus, malaria--many of these diseases seem to be a thing of the >past; but now we have more serious diseases such as AIDS, luekemia and >cancer at an all time HIGH.... > >I think Mendelsohn has made a significant point when stating that vaccines >are a medical time bomb. > >Thanks for the article ....although I cannot understand much of the >medical jargon. > >Lana >Mama of Cody Ukiah > > > > > >At 09:37 AM 9/6/98 -0600, you wrote: >>From: Mom2Q <Mom2Q@...> >> >> >>Sorry about the error. You have to be registered with Medscape to read >this article. Anyway here it is: >> >>The Search for Vaccines Against Helicobacter pylori >> >> P. Monath, MD, K. Lee, PhD, H. Ermak, PhD, Gwendolyn >A. Myers, DVM, >>A. Weltzin, PhD, J. sca, PhD, D. , Jr., PhD, >Gopalan Soman, PhD, Hitesh Bhagat, >>PhD, A. Ackerman, MD, Harry K. Kleanthous, PhD >> >>[infect Med 15(8):534-535,539-546, 1998. © 1998 SCP Communications, Inc.] >> >>Abstract >> >>Current antibiotic regimens against Helicobacter pylori are effective, but >complex dosing and development of resistance are >>concerns. Animal studies and limited clinical trials of H pylori urease >and other bacterial antigens have been conducted, with >>promising findings. [infect Med 15(8):534-535,539-546, 1998] >> >>Introduction >> >>One of the most promising recent developments in medicine is the concept >that chronic afflictions, such as peptic ulcer >>disease and cancer can be controlled through immunization like classic >infectious diseases. Research on vaccines against >>Helicobacter pylori-the leading cause of chronic gastritis and peptic >ulcer disease and a primary risk factor for gastric >>adenocarcinoma-began in 1990. The favored approach has been the oral >administration of purified recombinant subunit >>proteins of H pylori and a mucosal adjuvant, the labile toxin (LT) of >Escherichia coli. As a single-component vaccine, the >>urease protein has shown remarkable prophylactic and therapeutic activity >in animal models and partial therapeutic activity in >>humans. A number of other H pylori antigens have been effective in animal >models, and the recent sequencing of the >>complete H pylori genome has led to an intensive effort in antigen >discovery. Other research is directed at the comparison of >>adjuvants and vaccine delivery systems and toward the immunologic >mechanisms mediating protection. Here, we present >>preclinical data, the results of early-stage clinical trials, and >directions for future research on Helicobacter vaccines. >> >>Helicobacter pylori: Medical Impact >> >>A gram-negative spiral bacterium that specifically infects the stomach, H >pylori (Fig. 1) is one of the most prevalent >>infections of humankind: Approximately 50% of adults in the industrialized >world and more than 90% of inhabitants of >>developing countries are infected.[1] H pylori is thought to be acquired >by person-to-person spread via the fecal-oral and >>oral-oral routes, and in some areas it may be waterborne.[2] After oral >ingestion, the bacteria colonize gastric mucus in close >>association with gastric epithelial cells (Figs. 2,3). Infection is >chronic and generally lifelong. >> >>In the US, approximately 2.5 million new H pylori infections occur each >year. In Europe, the prevalence and incidence of H >>pylori-associated diseases are similar to or higher than those in the US. >In industrialized countries, the incidence of infection >>is decreasing overall, although transmission varies with socioeconomic >status, and subpopulations are thus differentially >>affected. >> >>H pylori is the cause of chronic gastritis and the vast majority of cases >of peptic ulcer disease.[3-6] Conclusive evidence also >>exists for an etiologic role of H pylori infection in dysplasia and >metaplasia of gastric mucosa, distal gastric adenocarcinoma, >>and non-Hodgkin's lymphoma of the stomach,[7-12] leading the World Health >Organization to classify the bacteria as a Class I >>(definite) carcinogen.[13] Considered in terms of lifetime morbidity, the >illness to infection ratio in the US and Europe may be >>estimated at 1:5 for peptic ulceration and 1:200 for gastric adenocarcinoma. >> >>Peptic Ulcer >> >>In the US, approximately 16,000 deaths are attributed annually to >complications of peptic ulcer disease. There are more >>than 2 million physician visits per year for duodenal ulcers, 90% of which >are attributable to H pylori, and more than 3 >>million physician visits per year for gastric ulcers, 60% of which are >attributable to the bacterium.[1,14] In a prospective study, >>the risk of developing duodenal ulcer disease in H pylori-infected >patients followed for 10 years exceeded 10%; in contrast, >>it was less than 1% in uninfected patients.[15] >> >>Gastric Adenocarcinoma >> >>The incidence of gastric adenocarcinoma in the US is approximately 24,000 >cases per year, with 13,300 deaths, >>approximately 60% of which (14,400 cases; 7980 deaths) may be attributed >to H pylori. The risk of developing gastric >>cancer is estimated to be 3- to 6-fold higher in infected than in >uninfected individuals.[7,12,16] Gastric cancer is a leading cause >>of death in Latin America and Asia. Acquisition of H pylori infection >early in life appears to be associated with early-onset >>gastric corpus atrophy and metaplasia and a higher risk of cancer.[17] >Ingestion of dietary carcinogens and deficiencies in >>dietary antioxidants are thought to be important cofactors in the genesis >of Helicobacter-related cancer. Helicobacter strain >>differences in virulence factors also appear to determine cancer risk.[18] >> >>Rationale for Vaccine Development >> >>Several lines of evidence provide a rationale for the development of a >vaccine against H pylori. >> >>High Illness to Infection Ratio >> >>Compared with illness to infection ratios of other infectious diseases, >that of H pylori-associated peptic ulcer is high (1:5). In >>comparison, such ratios are about 1:25 for hepatitis B-associated chronic >liver disease and about 1:10 for Mycobacterium >>tuberculosis. Although the ratio for H pylori-associated gastric >adenocarcinoma is lower (1:200), the morbidity and >>mortality associated with this disease are substantial. >> >>Vaccines for Prevention >> >>Vaccines have long been regarded as the most effective and economical >approach to the prevention and control of infectious >>diseases. Although effective antimicrobial treatment for H pylori is now >employed widely to prevent recurrence in patients >>with active or recent duodenal ulcer disease, the ability to treat does >not obviate the need for preventive strategies. Indeed, >>most H pylori infections leading to gastric cancer and 20% to 30% of cases >of upper gastrointestinal hemorrhage occur in >>individuals who have sustained long-term infections without antecedent >symptoms. For this reason, these individuals do not >>present to the physician in time for antimicrobial intervention. >> >>Vaccines as Therapy or Complement >> >>Although the application of vaccines for therapy of infectious diseases is >in its infancy, it has tremendous implications for the >>management of chronic infections, such as H pylori, HIV, human >papillomavirus, viral hepatitis, herpesviruses, chlamydia, >>and a wide range of parasitic infections. >> >>>From a practical perspective, the effectiveness of conventional >antimicrobials has diminished interest in therapeutic vaccines. >>However, vaccines used in combination with antibiotics could improve the >rate of treatment success and decrease the >>evolution of antimicrobial resistance and disease recurrence. >Infection-induced immunity to H pylori is clearly insufficient to >>prevent reinfection, as shown by experiments in animals[19] and limited >studies of humans.[20] In areas of the world with high >>rates of transmission of H pylori, re-infection may occur rapidly after >treatment with antimicrobial agents. However, in >>industrialized nations, re-infection rates in adults appear to be low, >both overall (0.5%-2%) and in high-risk individuals >>(2.5% in spouses of infected persons).[21] However, even in industrialized >nations, re-infection rates in children may be >>substantially higher; in 1 study, Oderda and colleagues[22] reported that >18% of children became re-infected within 18 >>months of antibiotic therapy. >> >>Convenient Identification ofH pylori >> >>A wide array of simple office-based serologic screening tests and the >noninvasive[13]C-urea breath test are now available for >>identifying infected individuals, and new serologic tests that identify H >pylori strains characterized by a higher virulence >>phenotype, especially CagA, are under development.[23] These methods could >be used to identify persons with H pylori >>gastritis during the first 2 decades of life, thus identifying a >population at future risk of ulcer disease and cancer. If treatment >>of the infection is considered, coadministration of a vaccine to prevent >re-infection will be an important component of such a >>strategy. >> >>Cost-effectiveness ofH pylori Vaccines >> >>Although there is convincing evidence for the cost-effectiveness of curing >H pylori in patients with duodenal ulcer disease,[24] >>the pharmacoeconomics of prophylactic immunization-whether primary >(pre-exposure) immunization or immunization to >>prevent re-infection-have not been well defined. Pre-exposure immunization >requires application during infancy or >>childhood, depending on age of acquisition of infection in the population >at risk. Since the indication for H pylori vaccination >>is the prevention of chronic diseases that occur in the third to the sixth >decade of life, the cost-benefit ratio is influenced by >>heavy discounting of future cost savings from disease prevention. However, >childhood immunization to prevent chronic >>disease acquired decades later is not without precedent and underlies the >recommendation for universal immunization against >>hepatitis B,[25] a disease that causes considerably less cancer morbidity >and mortality than H pylori.[26] In areas where gastric >>cancer is a leading cause of death, such as Latin America and Asia, >individuals and society place a high value on investments >>that reduce the incidence of this incurable and fatal disease. The World >Health Organization estimates that 550,000 gastric >>cancer deaths due to H pylori occur annually,[13] and these deaths must be >considered potentially preventable through >>immunization. By way of comparison, 316,000 cases of hepatocellular >carcinoma caused by hepatitis B occur annually, and >>many countries are implementing routine childhood immunization policies. >> >>Initial Immunization Trials >> >>Serious consideration of vaccination as a means to control peptic ulcer >disease began around 1990. Pallen and Clayton[27] >>suggested that urease would be a candidate antigen for incorporation in an >H pylori vaccine, based in part on findings in >>animals and humans immunized with jack-bean urease to suppress ammonia >production in the intestine by ureolytic bacteria. >>Czinn and Nedrud[28] showed that H pylori whole-cell sonicates >administered intragastrically to mice and ferrets elicited >>serum and intestinal immunoglobulin (Ig) G and IgA antibodies. Subsequent >studies by Chen and coworkers[29,30] and Czinn >>and colleagues[31] demonstrated that mice orally immunized with >Helicobacter sonicates or whole cells and cholera toxin >>(CT) adjuvant were protected against challenge with Helicobacter felis, a >species capable of infecting murine gastric >>mucosa. In addition, passive protection against challenge was demonstrated >by the oral administration of an IgA monoclonal >>antibody, suggesting that the principal mediator of protection after >active immunization may be secretory IgA. The protective >>monoclonal antibody later was shown to be specific for Helicobacter >urease.[32] >> >>In 1994, Michetti and others[33] demonstrated that mice orally immunized >with recombinant H pylori urease were protected >>against challenge with H felis. Protective determinants were present on >both subunits (UreA and UreB) of the recombinant >>multimeric urease molecule. The recombinant protein is similar to native >urease in multimeric structure, molecular mass >>(550kDa), and nano-particulate morphology.[34] The UreB subunit truncated >at the amino terminus,[35] and multimeric urease >>that had been aggregated or heat-denatured retained prophylactic activity >(OraVax, unpublished data, 1997). These studies >>clearly demonstrated that urease is remarkable among bacterial proteins in >its stability and immunogenicity. >> >>A large body of data has now been accumulated from several laboratories >confirming that H pylori urease administered >>mucosally to a variety of animals confers protection against >challenge.[34,36,37] While initial immunization studies utilized H felis >>as the challenge bacterium, the subsequent development of mouse models of >H pylori infection led to the confirmation that >>urease protected against the human pathogen.[38-40] >> >>In 1994, Doidge and colleagues[41] reported that mice with subchronic H >felis infection cleared or had reduced infection >>after oral immunization with H felis whole-cell sonicates. Urease >administered orally to mice experimentally infected with H >>felis[42] or ferrets naturally infected with Helicobacter mustelae[43] was >shown to have significant therapeutic activity. These >>studies indicated that the up regulation of immunity to specific H pylori >antigens may result in clearance of chronic infection. >>The role of mucosal immunity in protection against H pylori in humans is >also supported by a study of infants in West Africa, >>where infection usually occurs within the first year of life. Infants of >mothers with high titers of anti-Helicobacter IgA in >>breast milk had a significant delay in acquisition of H pylori >infection.[44] Subsequent studies indicate that the principal antigen >>recognized by breast milk IgA is urease (J. , MD, The Royal >n Infirmary, Newcastle Upon Tyne, England, >>personal communication, 1996). >> >>Approaches to Vaccine Development >> >>Although the feasibility of prophylactic and therapeutic immunization was >established by these initial studies, procedures for >>the large-scale production of a safe and effective product are needed >(Table I). The use of whole bacterial cells or cellular >>extracts is problematic, and while recombinant subunit vaccines >(especially urease) are attractive alternatives, the >>identification of a full complement of protective antigens to be included >in a recombinant vaccine remains a considerable >>challenge. However, the greatest problem for vaccine developers is the >selection of an effective method for presenting >>antigens to the host's immune system in such a way that protective or >therapeutic immune responses are elicited in the gastric >>mucosa. Since the mechanisms by which H pylori evades immunity and the >roles of T and B cells in effector responses are >>poorly understood, purely empirical approaches have been applied to screen >antigens, adjuvants, and delivery systems. >>Approaches using live H pylori strains, live vectors, and subunit antigens >have also been explored. >> >>LiveH pylori Vaccines >> >>Effective live, attenuated oral vaccines have been developed to protect >against several enteric bacterial infections, including >>typhoid, cholera, and Shigella. However, this approach poses certain >serious difficulties in the case of H pylori: >> >> Immunity resulting from infection with wild strains of H pylori does >not result in clearance or provide protection >> against superinfection with other H pylori strains, recrudescence >after antibiotic suppression, or re-infection after >> successful cure. A live, attenuated vaccine would probably elicit an >even weaker immune response than the wild-type >> bacteria. Thus, it would be technically difficult to modify H pylori >to induce effective immunity rather than the evasion >> or down-regulation of immunity associated with natural infection. >> >> It is likely that a live vaccine would require high doses (possibly >>/=109 organisms) and repeated administrations to be >> effective. Therefore, high-yield fermentation of H pylori is >difficult and may not be economically feasible at the scale >> required for a live vaccine. >> >> H pylori is well adapted to cause chronic, persistent infection in >the host. Since human host responses are highly >> variable and uncontrollable, an attenuated vaccine must not cause >persistent infection associated with an inflammatory >> response. Regulatory concerns about chronic infection with a vaccine >strain would require long-term follow-up studies >> in large populations. The sensitivity of tests for persistence of a >vaccine strain versus wild-type strains in humans is >> highly problematic. >> >> A live vaccine would elicit immune responses against a wide range of >antigens, some of which may be undesirable, >> due to cross-reactivity with homologous human antigens or stimulation >of delayed-type hypersensitivity responses. >> >> A live vaccine might be used as prophylaxis, but it is difficult to >conceive of its use for treatment of infection. >> >>Despite these concerns, there may be a role for a live, attenuated H >pylori vaccine in an effective prophylactic immunizing >>regimen. Preclinical studies in mice have demonstrated that H >pylori-specific T and B cells are recruited to the gastric >>mucosa in large numbers only after Helicobacter challenge.[45] In mice >immunized with urease before challenge, the gastric >>immune response is effective in clearing most of the challenge organisms, >but without the stimulus provided by the challenge, >>the stomach remains immunologically silent. This observation suggests that >an effective immunization might include priming of >>intestinal immunity with a subunit antigen, followed by a live, attenuated >H pylori vaccine that would direct the immune >>response to the gastric mucosa but would establish only a transient >infection sufficient to target immunity. The sequence of >>artificial immunizations in such a model may result in an immune response >that is qualitatively distinct from natural infection. >>This concept is currently being explored in our laboratories. >> >>Live Vectors >> >>Recombinant enteric bacterial vectors have been constructed to deliver >foreign antigens. Examples include attenuated strains >>of Shigella flexneri, Salmonella typhi, and E coli.[46] These vectors, as >well as others that replicate in the gastrointestinal >>tract or invade the body by this route, provide potential approaches to >immunization against mucosal pathogens such as H >>pylori. Examples of such vectors include Vibrio cholerae, Lactobacillus >species, Streptococcus gordonii, poxvirus, >>adenovirus, poliovirus, rhinovirus, and alphavirus. The ideal live vector >is one that is not replication-deficient or restricted in >>its ability to express its own and foreign antigenic determinants. >Restriction of vector replication by anti-vector immunity is a >>concern that can potentially be addressed by a combination of 2 >antigenically distinct vectors or a combination of parenteral >>priming followed by a live-vector boost or vice versa. Live vectors may >preclude the need for a mucosal adjuvant by >>targeting M cells and inductive lymphoid tissues in the gut. >Alternatively, the vectors may be designed to co-express antigens >>with immunomodulatory lymphokines. The use of live vectors could also >simplify vaccine administration schedules, since >>fewer doses would be required than of a subunit vaccine. The manufacturing >process is also greatly simplified, since protein >>purification is unnecessary. >> >>Preliminary studies have been performed in several laboratories with mixed >results, and it is too early to draw conclusions >>about the value of live vectors for construction of an effective >Helicobacter vaccine. >> >>Subunit Antigens >> >>Nonliving vaccines include defined subunits, whole-cell or crude >preparations, and DNA-based vaccines. Whole-cell or >>crude preparations appear to be effective in animal models and have the >advantage of multiple antigens presenting to the host >>without having to isolate, characterize, and prepare individually active >components. This approach is unlikely to be practical >>from a scale-up perspective or desirable from a regulatory view, given the >potential problem of autoimmunity due to >>Helicobacter antigens, such as blood group antigens (cross-reactive >with human cells).[47,48] DNA-based approaches >>are being investigated, but it is too early to assess the feasibility of >generating an effective mucosal (and especially gastric) >>immune response by this method. >> >>A nearer-term approach is the delivery of defined H pylori protein >antigens in a formulation designed to elicit protective >>responses in the stomach. H pylori bacteria have a number of virulence >factors that are of known importance in chronic >>infection, recruitment of inflammatory cells, and damage to mucosal >epithelium (Fig. 1). Among these, prominent is the >>urease enzyme, which is implicated in acid tolerance of the bacteria, >colonization, and mucin depletion. As noted, >>recombinant urease has been demonstrated to be highly effective in >prophylactic immunization of mice against challenge with >>Helicobacter species.[33-41] Evidence of protection has also been obtained >in models using larger animals, including cats and >>nonhuman primates.[49-51] >> >>Native urease is a metalloenzyme, dependent for enzymatic activity on >Ni2+, incorporated during intracellular synthesis.[50] >>Urease is essential for colonization of the stomach by H pylori; the >enzyme splits urea present in gastric juice to form >>ammonia, a strong base that presumably protects the bacterium from >inactivation by gastric acid.[51,52] All strains of H pylori >>that infect humans express the urease enzyme. In fact, urease accounts for >more than 6% of the total soluble bacterial protein >>of H pylori and is localized, in part, on the surface of the >bacterium.[50,53,54] This makes the urease enzyme an important >>target for the immune response elicited by a vaccine. Urease is >constitutively expressed in vivo so that the bacteria would be >>exposed to the anti-urease immune response during the entire course of >infection. Moreover, H pylori urease is intrinsically >>acid-stable, making it an ideal vaccine for oral application. H pylori >urease is highly conserved at the amino acid sequence >>level, and antigenic variation between strains of H pylori urease is not >likely to impair vaccine efficacy. Cross-reactivity >>between the ureases of different H pylori clinical isolates and between H >pylori urease and heterologous ureases of H felis >>and H mustelae has been demonstrated[52] and is the basis for the >heterologous cross-protection studies.[31] In its native >>form, urease is a hexameric structure of large molecular mass (550kDa), >composed of 6 copies of the UreA (30kDa) and >>UreB (60kDa) and has a particulate structure of 12nm in >diameter,[49,50,55] favoring uptake by M cells in the gastrointestinal >>tract for induction of mucosal immunity.[56] >> >>The vaccine candidate-recombinant urease-is urease antigen produced in >genetically engineered E coli. Antigenically >>indistinguishable from native urease, recombinant urease has an identical >particulate structure but is enzymatically >>nonfunctional and does not generate toxic ammonia in the presence of urea. >This has been accomplished by cloning and >>expressing in E coli only the genes for the structural subunits (ureA and >ureB), omitting all other genes of the operon,[57] and >>including those involved in insertion of Ni2+ required for enzymatic >function. After expression in fermentation cultures of E >>coli, the recombinant antigen is purified from bacterial lysates and is >subsequently lyophilized in a stabilizer. >> >>Therapeutic Immunization >> >>Treatment of H pylori infection in patients with peptic ulcer disease is >now an accepted health practice in the US[6] and >>Europe and is the basis for regulatory labeling of >antibiotic-antisecretory drug combinations. However, antimicrobial therapy >>has a number of inherent limitations that might be overcome by use of an >effective vaccine or a combined regimen of >>antibiotics and vaccine. On average, primary treatment failures occur in >15% of patients treated with antibiotics combined >>with an antisecretory drug. Poor compliance with complex antibiotic >regimens and antibiotic resistance in H pylori[58-60] >>contribute to treatment failures. In contrast to antibiotics, >vaccine-induced immunity is not expected to select for resistant or >>more virulent organisms. Since immunologic mechanisms are distinct from >those involved in antimicrobial treatment, vaccines >>alone or synergistic activities of vaccines and antimicrobials could >achieve the ultimate goal of 100% cure. >> >>Murine Studies >> >>Using recombinant urease[42,61] and crude cell antigens,[41] therapeutic >activity has been documented in mice, with efficacy >>rates (determined by gastric urease activity) between 50% and 94%. When >vaccine and a partially effective antibiotic >>regimen were combined, the latter proved to be more effective than either >treatment alone.[62] These studies were conducted >>in mice with subchronic H felis infection, the immunization regimen being >applied only a few weeks after infecting the >>animals. It is uncertain whether treatment would be as effective in a >chronically infected host. Moreover, the reported cure >>rates based on gastric urease or histologic endpoints overestimate the >effectiveness of immunization. In addition, in the >>mouse model, H felis is easier to eradicate than H pylori. The results >with vaccine are also supported by the observation >>that mice can be cured of H felis with a single antibiotic,[63] whereas >multiple drugs were required to achieve partial cure of >>H pylori.[64] When the H pylori mouse model was employed and therapeutic >activity of urease-LT immunization was >>measured by quantitative culture, a statistically significant (P = 0.0016) >10-fold reduction in bacterial density (not eradication >>of infection) was observed. Interestingly, the LT adjuvant alone appeared >to have some effect in reducing infection, possibly >>due to modulation of the immune response to antigens associated with >natural infection. >> >>Ferret Studies >> >>In ferrets, immunization with urease and CT adjuvant resulted in >presumptive cure of chronic H mustelae infection.[43] When >>tested 6 weeks after immunization, 30% of the ferrets were cured of >infection. A significant reduction in gastric inflammation >>was demonstrated by histopathology in up to 60% of the animals. >Interestingly, gastric inflammation was significantly >>reduced in the cured and persistently infected vaccinated animals compared >with infected controls, a finding similar to that >>described in the rhesus monkeys.[72] The possibility that vaccines can >diminish the pathologic consequences of Helicobacter >>infections deserves further study. >> >>Adjuvants >> >>All preclinical studies reported to date have demonstrated efficacy of >vaccination against Helicobacter infection, using >>antigens given mucosally together with CT or LT as a mucosal adjuvant. No >protection was achieved when antigens were >>administered without a mucosal adjuvant, even at exceedingly high >levels.[34] CT is not acceptable as a human adjuvant >>because it induces diarrhea in humans at microgram levels.[65] LT is less >reactogenic and has been tested clinically.[66] A >>possible means to circumvent the reactogenicity of native toxins as >adjuvants is the use of atoxic cholera toxin B subunit >>(CTB) spiked with a low dose of native toxin. This combination was shown >to be an effective adjuvant for an H felis >>sonicate vaccine, providing protection against H felis challenge.[34] An >even more attractive approach is the use of genetically >>detoxified LT molecules, which are enzymatically inactive but still retain >adjuvanticity.[67,68] >> >>Many novel adjuvants have shown promise in preclinical studies with a >variety of other vaccines, including oil emulsions, >>saponins, immunostimulating complexes, polyphosphazine, muramyl dipeptide >derivatives, block polymers, vitamin D3, >>liposomes, copolymer microspheres, and cytokines. Some data are now >available from clinical trials; more is known about >>many of these adjuvants for parenteral than for mucosal routes of >administration. In studies of H pylori urease antigen, a >>muramyl dipeptide derivative >(N-acetylglucosaminyl-N-acetyl-muramyl-L-alanyl-D-isoglutamine, GMDP) >delivered orally >>did not elicit protection in mice against challenge with H felis.[34] Alum >given parenterally with urease was partially effective >>when given prophylactically (OraVax, unpublished data, 1997). An >exploration of various adjuvants for parenteral >>immunization with urease and for combined mucosal-parenteral immunization >regimens is currently underway in our >>laboratory and that of our partner, Pasteur Merieux Connaught. Preliminary >data indicate that partial protection is achieved >>by parenteral injection of antigen with alum and other select adjuvants. >Since adjuvants orient the immune response in a >>selective fashion with respect to T-helper subsets, the results of >comparative studies will shed light on the role of Th1 and >>Th2 responses in protection. Immunization studies of interleukin-4 >knock-out and gamma-interferon receptor deficient mice >>indicate that both Th1 and Th2 responses are required for protective >immunity.[69] This finding is also supported by our >>observations of adjuvants having selective immunomodulatory properties. >> >>Clinical Trials >> >>Clinical testing of recombinant urease was initiated by our group in 1994, >and trials of whole-cell and other recombinant >>antigens are in the planning stages by others. Our clinical studies were >begun in healthy infected volunteers (rather than >>uninfected subjects) because of concern that immunization of naive >individuals may potentiate inflammation upon subsequent >>infection. This phenomenon was at that time observed in mice[33,42,70] but >subsequently not observed in cats or monkeys. In >>addition, because the immune correlates of protection remain problematic, >it was believed that the direct measurement of a >>therapeutic effect in infected subjects would have the greatest clinical >significance. >> >>A limited study was first performed to demonstrate the safety and >tolerability of oral administration of urease without a >>mucosal adjuvant.[71] In a randomized, double-blind, placebo-controlled >trial conducted by Kreiss and colleagues,[71] 6 >>infected asymptomatic adults were administered 4 doses of vaccine-each >consisting of 60mg of recombinant H pylori >>urease-by the oral route once a week. Six infected subjects received >placebo. As expected in the absence of an adjuvant, >>none of the vaccinated individuals mounted an immune response, and in >gastric biopsies obtained before and 1 month after >>vaccination, no change in bacterial density (measured by quantitative >culture), inflammation, or mucosal damage was >>observed. No adverse events was attributable to administration of urease. >> >>A second trial was conducted to determine the tolerability of >coadministration of urease with a mucosal adjuvant (LT) in >>healthy adults with H pylori infection and to obtain preliminary data on >therapeutic activity. Preliminary results of this >>trial-which was conducted at the Centre Hospitalier Universitaire, >Lausanne, and at the Center for Vaccine Development, >>University of land in Baltimore-were reported by Michetti and >colleagues at the Helicobacter congress in >>Copenhagen in 1996. Native LT purified from E coli was supplied by the >Naval Medical Research Institute in Bethesda, >>land, which previously reported adjuvant activity in a study involving >cholera vaccine.[66] >> >>The controlled trial involved administration of 4 weekly, graded doses of >urease (20, 60, or 180mg) with LT; placebo >>vaccine with LT; or placebo vaccine and placebo adjuvant to groups of 4 or >5 volunteers. The ELISPOT assay for >>antibody-secreting cells (ASCs) in peripheral blood was the most sensitive >determinant of immunologic response to the >>vaccine. Six of 14 (43%) subjects who received urease, but none of the 10 >subjects who received placebo vaccine, had an >>increase in IgA or IgG ASCs at 1 or more time points, measured 7 days >after each successive dose of vaccine. Gastric >>biopsies were obtained before and 1 month after completion of the >immunization regimen. Differences were determined >>between pre- and postimmunization H pylori densities in gastric mucosa. >Pairwise treatment group comparisons were >>performed at baseline and on the change from baseline to postimmunization. >In addition, the significance of the mean change >>from baseline to postimmunization was assessed within each treatment >group. While the urease-treated groups were not >>significantly different from control groups with respect to the change >from baseline to postimmunization, the subjects >>receiving active urease experienced, on average, a larger decrease in >bacterial densities from baseline to postimmunization >>(P = 0.032) than did subjects receiving placebo (P = 0.425). While the >study had small sample sizes per group and was not >>powered to detect significant differences between treatment groups, it >provided the first clinical evidence for a therapeutic >>activity of oral urease with LT adjuvant. The duration of the study was >not sufficient to assess whether administration of the >>vaccine was associated with a decrease in inflammation, as has been >observed in ferrets[43] and rhesus monkeys.[72] >> >>Conclusions and Future Research >> >>A convincing body of data now exists supporting the potential for >successful immunization against H pylori. However, we >>are still at a preliminary stage in clinical development. The best >immunogens, the best mode of presentation, the number of >>doses needed, optimal age at immunization, expected benefit, >cost-effectiveness, and other factors involved in vaccine >>development require further study. >> >>The complex pathogenesis of this infection,[3,73] including the presence >of antigens on H pylori shared with the host (a >>mechanism for immune evasion),[48] demands novel approaches to the >development of a final vaccine formulation. The >>selection of defined and well-characterized recombinant subunit antigens >appears to be the most viable approach, and the >>urease antigen has so far proved most potent in eliciting protective >immunity. It is reasonable to assume that more than 1 >>protective component is needed in a vaccine, and a number of such antigens >in addition to urease have now been >>discovered. The sequence analysis of the entire H pylori genome by Tomb >and colleagues[74] will enhance antigen discovery >>efforts. In addition to antigen composition, a successful vaccine must be >delivered to the host in a manner that elicits >>protective (therapeutic) immunity, particularly immunity expressed at the >site of bacterial colonization (gastric mucosa). The >>most appropriate means to achieve this end has not yet been fully defined. >Mucosal routes of immunization with a classic >>mucosal adjuvant (LT) have yielded the best results, but prophylactic >(therapeutic) activity remains incomplete. Research is >>needed on the mechanisms of protective immunity induced by vaccines, on >the protein-specific immune responses to natural >>infection, and on the functional role of T cells. Such studies may provide >important data that lead to novel immunization >>methods, as well as surrogate tests for protection that are useful in >vaccine trials. >> >>Additional discussion of animal models for the development of Helicobacter >vaccine can be found on Medscape >>(www.medscape.com). >> >>Acknowledgments >> >>Original work described in this paper was funded in part by Pasteur >Merieux Connaught (PMC) and by the National >>Institutes of Health. The authors are grateful to PMC scientists, >particularly Drs. Pierre Meulien, Marie- Quentin-Millet, >>Farukh Rizvi, Bruno Guy, Ling Lissolo, and Veronique Mazarin (PMC, Marcy >l'Etoile, France) for their scientific input >>about the work and its interpretation. Drs. Pierre Michetti, Christiana >Kreiss, Irene Couthesy-Theulaz, and Andre Blum >>(Centre Hospitalier Universitaire, Lausanne, Switzerland); Kotloff >and Genevieve Losonsky (Center for Vaccine >>Development, University of land, Baltimore, Md.); and >(University of land Medical Center, >>Baltimore, Md.) conducted the clinical trials reviewed in this paper. Drs. > Czinn and Nedrud (Case-Western >>Reserve University, Cleveland, Ohio); Fox (Massachusetts Institute >of Technology, Cambridge, Mass.); Andre >>Dubois (Uniformed Services University of the Health Sciences, Bethesda, >Md.); Soike (Tulane University, >>Covington, La.); and ph Hill, Christian Stadtlander, Hal Farris, and > Gangemi (Clemson University, Clemson, >>S.C.) made significant contributions in many aspects of the testing of >Helicobacter vaccine candidates in animal models. The >>authors are especially grateful for the excellent assistance of OraVax >personnel, including ph Simon, Kochi, >> Tibbitts, Ingrassia, Gray, Kathleen >Georgokopoulos, Amal Al-Gawari, , Rue Ding, >>and Bruce Ekstein. >> >>References >> >> 1. DN, Parsonnet J: Epidemiology and natural history of >Helicobacter pylori infection, in Blaser MJ, PD, >> Ravdin J (eds): Infections of the Gastrointestinal Tract. New York, >Raven Press, 1994. >> >> 2.Klein PD, Graham DY, Gaillour A, et al: Water source as a risk factor >for Helicobacter pylori infection in Peruvian >> children. Lancet 337:1503-1506, 1991. >> >> 3.Blaser MJ: Helicobacter pylori and the pathogenesis of gastroduodenal >inflammation. J Infect Dis 161:626-633, >> 1990. >> >> 4.Cover TL, Blaser MJ: Helicobacter pylori and gastroduodenal disease. >Annu Rev Med 43:135-145, 1992. >> >> 5.Solnick JV, Tompkins LS: Helicobacter pylori and gastroduodenal >disease: Pathogenesis and host-parasite >> interaction. Infect Agents Dis 1:294-309, 1993. >> >> 6.National Institutes of Health: Consensus development conference >statement: Helicobacter pylori in peptic ulcer >> disease. JAMA 272:65-69, 1994. >> >> 7.Eurogast Study Group: An international association between >Helicobacter pylori infection and gastric cancer. Lancet >> 341:1359-1362, 1993. >> >> 8.Forman D, Webb P: Geographic distribution and association with >gastric cancer, in Northfield TC, Mendall M, >> Goggin PM (eds): Helicobacter pylori Infection. Dordrecht, Germany, >Kluwer Academic Publishers, 1993, pp >> 11-20. >> >> 9.Correa P: Human gastric carcinogenesis: A multistep and >multifactorial process. Cancer Res 52:6735-6740, 1992. >> >> 10.Wotherspoon AC, Doglioni C, Diss TC, et al: Regression of primary >low-grade B-cell gastric lymphoma of >> mucosa-associated lymphoid tissue type after eradication of >Helicobacter pylori. Lancet 342:575-577, 1993. >> >> 11.Parsonnet J, Hansen S, L, et al: Helicobacter pylori >infection and gastric lymphoma. N Engl J Med >> 18:1267-1271, 1994. >> >> 12.Parsonnet J, Samloff IM, LM, et al: Helicobacter pylori, >pepsinogen and risk for gastric adenocarcinoma. >> Cancer Epidemiol Biomarkers Prev 2:1-7, 1993. >> >> 13.World Health Organization: The State of World Health. Geneva, World >Health Organization, 1996, pp 59-62. >> >> 14.Goodwin CS, Worsley BW: Helicobacter pylori: Biology and Clinical >Practice. Boca Raton, CRC Press, 1993, p >> 465. >> >> 15.Sipponen P: Natural history of gastritis and the relationship to >peptic ulcer disease. Digestion 5:70-75, 1990. >> >> 16. HM: The epidemiology of Helicobacter pylori infection and >its relation to gastric cancer, in Goodwin CS, >> Worsley BW (eds): Helicobacter pylori: Biology and Clinical Practice. >Boca Raton, CRC Press, 1993, pp 95-114. >> >> 17.Sipponen P, Seppala K: Gastric carcinoma: Failed adaptation to >Helicobacter pylori. Scand J Gastroenterol >> 27(suppl 1):S33-S38, 1992. >> >> 18.Blaser MJ, - G, Kleanthous H, et al: Infection with >Helicobacter pylori strains possessing cagA is >> associated with an increased risk of developing adenocarcinoma of the >stomach. Cancer Res 55:1-5, 1995. >> >> 19.Lee CK, Soike K, Tibbitts T, et al: Urease immunization protects >against re-infection by Helicobacter pylori in >> rhesus monkeys. Gut 39(2)(suppl):A45, 1996. >> >> 20.Gilman RH, Leon-Barua R, -Ramos A, et al: Rapid recurrence of >Helicobacter pylori infection in Peruvian >> patients after eradication therapy. Proc Clin Infect Dis J 1997 (in >press). >> >> 21.Marshall BJ: Epidemiology of H pylori in western countries, in Hunt >RH, Tytgat GNJ (eds): Helicobacter pylori: >> Basic Mechanisms Top Clinical Cure. Dordrecht, Germany, Kluwer >Academic Publishers, 1994, pp 75-84. >> >> 22.Oderda G, Vaira D, Ainley C: Eighteen month follow-up of H pylori >positive children treated with amoxicillin and >> tinidazole. Dig Dis Sci 36:572-576, 1992. >> >> 23.Cover TL, Glupczynski Y, Lage AP: Serologic detection of infection >with cagA+ Helicobacter pylori strains. J Clin >> Microbiol 33:1496-1500, 1995. >> >> 24.Deltenre M, Jonsson B: Pharmacoeconomics. Curr Opin Gastroenterol >12(suppl 1):S45-S49, 1996. >> >> 25.US Public Health Service: Vaccination and prophylaxis against >hepatitis B in children. Am Fam Physician >> 50:979-984, 1994. >> >> 26.US Public Health Service: Important Information About Hepatitis B, >Hepatitis B Vaccine, and Hepatitis B Immune >> Globulin. Washington, DC: US Depart Health Hum Services, May 1992. >> >> 27.Pallen MJ, Clayton CL: Vaccination against Helicobacter pylori >urease. Lancet 336:186-187, 1990. >> >> 28.Czinn SJ, Nedrud JG: Oral immunization against Helicobacter pylori. >Infect Immun 59:2359-2363, 1991. >> >> 29.Chen M, Lee A: Vaccination possibilities and probabilities, in >Northfield TC, Mandell M, Goggin PM (eds): >> Helicobacter pylori Infection. Dordrecht, Germany, Kluwer Academic >Publishers, 1993, pp 158-169. >> >> 30.Chen M, Lee A, Hazell S: Immunization against gastric Helicobacter >infection in a mouse Helicobacter felis model. >> Lancet 339:1120-1121, 1992. >> >> 31.Czinn SJ, Cai A, Nedrud JG: Protection of germ-free mice from >infection by Helicobacter felis after active oral or >> passive IgA immunization. Vaccine 11:637-642, 1993. >> >> 32.Blanchard TG, Czinn SJ, Maurer R, et al: Urease specific monoclonal >antibodies prevent Helicobacter felis infection >> in mice. 63:1394-1399, 1995. >> >> 33.Michetti P, Couthesy-Theulaz I, Davin C, et al: Immunization of >BALB/c mice against Helicobacter felis infection >> with Helicobacter pylori urease. Gastroenterology 107:1002-1011, 1994. >> >> 34.Lee CK, Weltzin RA, WD Jr., et al: Oral immunization with >recombinant Helicobacter pylori urease >> induces secretory IgA antibodies and protects mice from challenge >with Helicobacter felis. J Infect Dis >> 172:161-172, 1995. >> >> 35.Dore-Davin C, Michetti P, Saraga E, et al: A 37 kDa fragment of UreB >is sufficient to confer protection against >> Helicobacter felis infection in mice. Gastroenterology >110(suppl):A97, 1996. >> >> 36.Pappo J, WD Jr., Kabok Z, et al: Effect of oral immunization >with recombinant urease on murine >> Helicobacter felis gastritis. Infect Immun 63:1246-1252, 1995. >> >> 37.Ferrero RL, Thiberge JM, Huerre M, et al: Recombinant antigens >prepared from the urease subunits of Helicobacter >> spp.: Evidence of protection in a mouse model of gastric infection. >Infect Immun 62:4981-4989, 1994. >> >> 38.Marchetti M, Arico B, Burroni D, et al: Development of a mouse model >of Helicobacter pylori infection that mimics >> human disease. Science 267:1655-1658, 1995. >> >> 39.Kleanthous H, Tibbits T, Bakios TJ, et al: In vivo selection of a >highly adapted H pylori isolate and the development >> of an H pylori mouse model for studying vaccine efficacy. Gut >37(suppl):A94, 1995. >> >> 40.Kleanthous H, Myers G, Georgokopoulos K, et al: Effect of route of >mucosal immunization with recombinant urease >> on gastric immune responses and protection against Helicobacter >pylori infection. Gut 39(suppl 2):A75, 1996. >> >> 41.Doidge C, Gust I, Lee A, et al: Therapeutic immunization against >Helicobacter infection. Lancet 343:914-915, 1994. >> >> 42.Couthesy-Theulaz I, Porta N, Glauser M, et al: Oral immunization with >Helicobacter pylori urease B subunit as a >> treatment against Helicobacter infection in mice. Gastroenterology >109:115-121, 1995. >> >> 43.Cuenca R, Blanchard TG, Czinn SJ, et al: Therapeutic immunization >against Helicobacter mustelae in naturally >> infected ferrets. Gastroenterology 110:1770-1775, 1996. >> >> 44. JE, Austin S, Dale A, et al: Protection by human milk IgA >against Helicobacter pylori infection in infancy. >> Lancet 342:121, 1993. >> >> 45.Ermak TH, Kleanthous HK, Myers G, et al: Oral immunization of mice >with recombinant Helicobacter pylori urease: >> Corpus gastritis after challenge with H felis is due to residual >bacteria. Immunol Cell Biol 75 (suppl 1):A92, 1997. >> >> 46.Noriega FR, Losonsky G, Wang JY, et al: Further characterization of >aroA virG Shigella flexneri a strain >> CVD1203 as a mucosal Shigella vaccine and a live-vector vaccine for >delivering antigens of enterotoxigenic >> Escherichia coli. Infect Immun 64:23-27, 1996. >> >> 47.Negrini R, Savio A, Poiesi C, et al: Antigenic mimicry between >Helicobacter pylori and gastric mucosa in the >> pathogenesis of body atrophic gastritis. Gastroenterology >111:655-665, 1996. >> >> 48.Appelmelk BJ, Simoons-Smit I, Negrini R, et al: Potential role of >molecular mimicry between Helicobacter pylori >> lipopolysaccharide and host blood group antigens in >autoimmunity. Infect Immun 64:2031-2040, 1996. >> >> 49.Mobley HLT, Foxall PA: H pylori urease, in Hunt R, Tytgat G (eds): >Helicobacter pylori: Basic Mechanisms to >> Clinical Care. Dordrecht, Germany, Kluwer Academic Publishers, 1994, >pp 41-58. >> >> 50.Mobley HLT, Island MD, Hausinger RP: Molecular biology of microbial >ureases. Microbiol Rev 59:451-480, 1995. >> >> 51.Ferrero RL, Lee A: The importance of urease in acid protection for >the gastric-colonizing bacteria Helicobacter >> pylori and H felis sp nov. Microb Ecol Health Dis 4:121-134, 1991. >> >> 52.Hawtin PR, Stacey AR, Newell DG: Investigation of the structure and >localization of the urease of Helicobacter >> pylori using monoclonal antibodies. J Gen Microbiol 136:1995-2000, 1990. >> >> 53.Phadnis SH, Parlow MH, Levy M, et al: Surface localization of >Helicobacter pylori urease and a heat shock protein >> homologue requires bacterial autolysis. Infect Immun 64:905-912, 1996. >> >> 54.Gootz TD, - G, Clancy J, et al: Immunological and molecular >characterization of Helicobacter felis >> urease. Infect Immun 62:793-798, 1994. >> >> 55.Dunn BE, CP, - G, et al: Purification and >characterization of urease from Helicobacter pylori. J >> Biol Chem 265:9464-9469, 1990. >> >> 56.Neutra MR, Kraehenbuhl J-P: Transepithelial transport of proteins by >intestinal epithelial cells, in Audus KL, Raub TJ >> (eds): Biological Barriers to Protein Delivery. New York, Plenum >Press, 1993, pp 107-129. >> >> 57.Labigne A, Cussac V, Courcoux P: Shuttle cloning and nucleotide >sequence of Helicobacter pylori genes >> responsible for urease activity. J Bacteriol 173:1920-1931, 1991. >> >> 58.Noach LA, Langenberg WL, Bertola MA, et al: Impact of metronidazole >resistance on the eradication of >> Helicobacter pylori. Scand J Infect Dis 26:321-327, 1994. >> >> 59.Megraud F: Helicobacter pylori resistance to antibiotics, in Hunt RH, >Tytgat GNJ (eds): Helicobacter pylori: Basic >> Mechanisms to Clinical Cure. Dordrecht, Germany, Kluwer Academic >Publishers, 1994, pp 570-583. >> >> 60.Parasakthi N, Goh KL: Primary and acquired resistance to >clarithromycin among Helicobacter pylori strains in >> Malaysia. Am J Gastroenterol 90:519, 1995. >> >> 61.Kleanthous H, Ermak T, Pappo J, et al: Oral immunization with >recombinant Helicobacter pylori urease apoenzyme >> in the treatment of Helicobacter infection. Gut 37(suppl):A94, 1995. >> >> 62.Kleanthous H, Tibbitts T, Bakios J, et al: Oral immunization with >recombinant urease combined with antimicrobial >> therapy augment clearance of an H felis infection in mice. Gut >39(suppl 2):A75, 1996. >> >> 63.Hook-Nikanne J, Aho P, Karkkainen P, et al: The Helicobacter felis >mouse model in assessing anti-Helicobacter >> therapies and gastric mucosal prostaglandin E2 levels. Scand J >Gastroenterol 31:334-338, 1996. >> >> 64.Dubois A, Fiala N, Heman-Ackah LM, et al: Natural gastric infection >with Helicobacter pylori in monkeys: A model >> for human infection with spiral bacteria. Gastroenterology >106:1405-1417, 1994. >> >> 65.Levine MM, Black RE, Clements ML, et al: Volunteer studies in >development of vaccines against cholera and >> enterotoxigenic Escherichia coli, in Holme T, Holmgren J, Merson M, >et al (eds): Acute Enteric Infection in >> Children: New Prospects for Treatment and Prevention. Amsterdam, >Elsevier, 1981, pp 443-459. >> >> 66. DA, Baqar S, Oplinger M, et al: Safety and adjuvant activity of >native and mutant E coli heat-labile toxins. >> From Progress on Modulation of the Immune Response to Vaccine >Antigens, a symposium of the International >> Association of Biology Standards Task Force on Vaccines, World Health >Organization, Bergen, Norway, June >> 18-21, 1996. >> >> 67.Dickinson BL, Clements JD: Dissociation of Escherichia coli >heat-labile enterotoxin adjuvanticity from >> ADP-ribosyltransferase activity. Infect Immun 63:1617-1623, 1995. >> >> 68.Tommaso AD, Saletti G, Pizza M, et al: Induction of antigen-specific >antibodies in vaginal secretions by using a >> nontoxic mutant of heat-labile enterotoxin as a mucosal adjuvant. >Infect Immun 64:974-979, 1996. >> >> 69.Radcliff FJ, Ramsay AJ, Lee A: A mixed Th1/Th2 response may be >necessary for effective immunity against >> Helicobacter. Immunol Cell Biol 75(suppl 1):A90, 1997. >> >> 70.Ermak TH, Ding R, Ekstein B, et al: Oral immunization of mice with >recombinant Helicobacter pylori urease: Corpus >> gastritis after challenge with H felis is due to the presence of >residual bacteria. Gastroenterology 113:1118-1128, >> 1997. >> >> 71.Kreiss C, Buclin T, Cosma M, et al: Safety of oral immunization with >recombinant urease in patients with >> Helicobacter pylori infection. Lancet 347:1630-1631, 1996. >> >> 72.Dubois A, Lee CK, Fiala N, et al: Immunization against natural >Helicobacter pylori infection in rhesus monkeys. >> Gastroenterology, 1998. In press. >> >> 73.Labigne A, de Reuse H: Determinants of Helicobacter pylori >pathogenicity. Infect Agents Dis 5:191-202, 1996. >> >> 74.Tomb JF, White O, Kerlavage AR, et al: The complete genome sequence >of the gastric pathogen Helicobacter >> pylori. Nature 388(6642):539-547, 1997. >> >> >> >> >>------------------------------------------------------------------------ >> Quote Link to comment Share on other sites More sharing options...
Recommended Posts
Join the conversation
You are posting as a guest. If you have an account, sign in now to post with your account.
Note: Your post will require moderator approval before it will be visible.