AU675938C - Oral treatment of helicobacter infection - Google Patents

Oral treatment of helicobacter infection

Info

Publication number
AU675938C
AU675938C AU39770/93A AU3977093A AU675938C AU 675938 C AU675938 C AU 675938C AU 39770/93 A AU39770/93 A AU 39770/93A AU 3977093 A AU3977093 A AU 3977093A AU 675938 C AU675938 C AU 675938C
Authority
AU
Australia
Prior art keywords
felis
helicobacter
antigen
antibody
infection
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn - After Issue
Application number
AU39770/93A
Other versions
AU675938B2 (en
AU3977093A (en
Inventor
Steven James Czinn
John Gilbert Nedrud
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sanofi Pasteur Biologics LLC
Original Assignee
OraVax Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by OraVax Inc filed Critical OraVax Inc
Priority claimed from PCT/US1993/003409 external-priority patent/WO1993020843A1/en
Publication of AU3977093A publication Critical patent/AU3977093A/en
Publication of AU675938B2 publication Critical patent/AU675938B2/en
Application granted granted Critical
Publication of AU675938C publication Critical patent/AU675938C/en
Anticipated expiration legal-status Critical
Withdrawn - After Issue legal-status Critical Current

Links

Description

ORAL TREATMENT OF HELICOBACTER INFECTION
The present invention relates to the treatment of gastric infection in mammals, including humans. More particularly, the present invention relates to a method for the treatment of Helicobacter infection in mammals, including humans, and to vaccine compositions and antibodies suitable for use in such treatment.
BACKGROUND OF THE INVENTION
Helicobacter pylori (H. pylori) infection of human gastric epithelium is a major factor in the development of gastritis and ulcers and may be a risk factor for the development of gastric cancer 1-3
This slender S-shaped gram negative microorganism is routinely recovered from gastric tissue of adults and children with histologic evidence of gastritis or peptic ulceration. Evidence for a causal relationship between H. pylori and gastroduodenal disease comes from studies in human volunteers, gnotobiotic pigs, and germ-free rodents whereby postulates by Koch were satisfied by creating histologically confirmed gastritis following consumption of viable microorganisms 4-11. Although difficult to treat, when eradication is achieved the underlying gastritis resolves and, in patients with duodenal ulcer disease, the recurrence rate of the ulcer decreases dramatically 12
In spite of in vitro susceptibility to many antimicrobial agents, in vivo long-term eradication of established H. pylori infections with
SUBSTITUTESHEET antimicrobial agents is difficult to achieve18. The microorganism is found within the mucous coat overlying the gastric epithelium. This is a location which does not appear to allow for adequate antimicrobial levels to be achieved when given orally. At the present time, most authorities recommend a "triple therapy", namely a bismuth salt in combination with tetracycline and metronidazole for 2-4 weeks. However, the effectiveness of this or other chemotherapeutic regimens remains suboptimal.
At the present time little is known regarding the role of the mucosal immune system in the stomach.
The distribution of Ig producing cells in the normal gastric antrum indicates that IgA plasma cells make up 80% of the total plasma cell population. In addition, the number of plasma IgA cells present in the gastric antrum is comparable to other mucous membranes 25'26. Although a number of studies have looked at immunoglobulin levels in various endocrine fluids, no data is available regarding the concentration of immunoglobulins in gastric secretions. Moreover there is only limited data to _. suggest that patients infected with H. pylori develop specific IgG and/or IgA antibodies in gastric aspiarates 32. Thus, once infection is established, neither antibody nor antibiotics are very effective at eradication.
Czinn et al have shown that repetitive oral immunizations with H. pylori antigens and cholera toxin result in the inducement of a vigorous gastrointestinal IgA anti-H. pylori response in mice and ferrets 18. However, since mice and ferrel are. resistant to H. pylori infection and since no
SUBSTITUTE SHEET small animal model existed at that time to evaluate protection, it was unknown whether the antibodies so formed were protective.
. Lee et al have reported the ability to infect germ-free rodents with H. felis and reproducibly document histologic gastritis 9'10. However, no evaluation of protection has been reported.
There remains a need therefore for an effective treatment of H. pylori gastric infection, especially in humans. The present invention seeks to fill that need.
SUMMARY OF THE INVENTION
The present inventors have discovered, "surprisingly, that oral immunization of a host with Helicobacter antigen results in the formation of antibodies which are protective against acute infection by Helicobacter microorganisms. The formation of such protective anibodies was not predictable on the basis of prior work since, prior to the present invention, no suitable model existed to evaluate protection.
According to one aspect of the present invention, there is provided a method of eliciting in a mammalian host a protective immune response to Helicobacter infection, comprising orally administering to the host an immunogenically effective amount of Helicobacter antigen to elicit the desired protective immune response.
According to another aspect of the present invention, there is provided a vaccine composition comprising an amount of Helicobacter antigen
SUBSTITUTESHEET effective to elicit a protective human response in a patient, in association with a pharmaceutically acceptable diluent.
According to a further aspect of the present invention, there is provided a method of imparting to a mammalian host passive protection to Helicobacter infection, comprising orally administering to the host a immunologically effective amount of a Helicobacter specific IgA antibody to impart the desired passive protection.
According to yet another aspect of the present invention, there is provided a murine H. felis specific IgA or IgG monoclonal antibody.
According to a yet further aspect of the invention, there is provided a cell line #71-G5-A8.
BRIEF DESCRIPTION OF THE DRAWINGS
The invention will now be further described with reference to the accompanying figures, in which:
Figure 1 is is a bar chart of antibody titers in various sera and secretions of germ free mice after oral immunization with H. felis lysate in association with cholera toxin; and
Figures 2A and 2B are bar charts of percent of mice infected with H. felis after active immunization (Figure 2A) and passive immunization (2B) compared with controls.
DETAILED DESCRIPTION OF THE INVENTION
The present inventors have demonstated that oral
SUBSTITUTESHEET immunization in mice using H. felis antigen produces a protective immune response wherein antigen specific protective antibodies are present in gastric secretions. The effect of the protective immune response is that immunized animals when challenged with pathogen do not become infected in comparison to non-immunized animals which do become infected. While not being bound by any theory, the present inventors believe that oral immunization with the H. felis antigen stimulates the common mucosal immune system and perhaps local sites in the gastric mucosa resulting in the appearance of H. felis specific IgA antibodies in the gastric secretions, which prevent H. felis infection. Since H. felis and H. pylori are simliar species from the same genus (Helicobacter), it is reasonable to conclude that immunization of for example a germ-free pig with H. pylori antigen plus a mucosal adjuvant such as cholera toxin will be effective in preventing H. pylori infection of the stomach. Since it is a routine matter to conduct pre-clinical trials of candidate vaccines for human use in animal models, t is believed that the methodology of the present invention is effective in humans, especially in the treatment of H. pylori infection in humans.
It has been discovered by the present inventors that an H. felis germ-free mouse model can be employed to evaluate antibody protection levels following immunization with H. felis antigen. Figure 1 relates to the results obtained in experiments with the H. felis germ-free mouse model. Oral immunization of the model with bacterial antigens in association with cholera toxin resulted
SUBSTITUTESHEET in elevated serum, gastric and intestinal anti-H. felis antibody titers and protection from acute infection of the stomach by H. felis pathogen. In the experiments, groups of Swiss-Webster germ-free mice (Taconic) were orally immunized 4 or 5 times over a one month period with 2-4 mg of sonicated H. felis lysate plus 10 μg of cholera toxin. The mice were then challenged orally with approximately 10 viable H. felis bacteria. The mice were sacrificed and intestinal and gastric secretions collected as described in the following working Examples. Anti-H. felis antibody titers were determined by ELISA. The black solid bars in Figure 1 represent mean titers (± S.D.) from immunized mice and the open bars represent mean titers (± S.D.) from the control non-immunized mice. The results presented graphically in Figure 1 are summarized in Table 1 below.
TABLE 1
ANTIBODY TITER ( 0G2)
Serum Gastric Intestine IgA IgG IgA IgG IgA IgG
CONTROL 3.1 3 0 0 1.6 1.1 IMMUNIZED 11.8 16.8 2.1 4.25 4.5 4.4
SUBSTITUTE SHEET It can be seen from the above results that significantly higher antibody titers are observed for the immunized mice than for the control animals.
Figures 2A and 2B depict the results of studies to establish the protection against infection by H. felis by conducting active and passive immunization experiments. Referring to the active immunization experiments, gastric biopsies were collected at sacrifice from the H. felis challenged mice in the experiments described above in connection with Figure 1. The biopsies were scored for the presence of H. felis by rapid urease test and/or culture positivity, described in the following working Examples. Figure 2A shows the results of pooled data from 3 experiments (n = 17 immunized animals and 18 control animals). The black (solid) bars represent challenged immunized mice and the striped bars the control non-immunized mice.
It will be seen that from a total of 17 immunized animals, only 4 became infected, as compared to 14 of the 18 control animals. In other words, 78% percent of the immunized animals were protected from H. felis infection as compared to 23% of the non-immunized animals.
~ The fact that protection was the direct result of IgA antibodies was established by passive immunization of germ-free mice with H. felis specific IgA monoclonal antibodies and comparison of the resulting protection with that exhibited by mice given no antibody or irrelevant antibody (for example Sendai virus specific IgA monoclonal antibody) . The results are set forth in Figure 2B.
An IgA monoclonal antibody reactive with H.
SUBSTITUTESHEET felis was isolated and εubcloned after an immunization protocol similar to that described in Figure 1. Ascites containing H. felis specific IgA monoclonal antibody produced from the cell line #71-G5-Aβ, prepared as described in the working Examples, or Sendai virus specific IgA monoclonal antibody or saline were orally administered to germ-free mice at the time of infection with H. felis, and 4, 8, and 24 hours later. Seven days after infection, the mice were sacrificed and gastric biopsies scored for H. felis (n = 7 mice received H. felis specific monoclonal antibody and 13 mice received no antibody or Sendai virus specific monoclonal antibody) . The black sold bars represent the mice which received the H. felis specific monoclonal antibody and the striped bars represent the mice which received either Sendai virus specific monoclonal antibody or saline (no antibody) .
These results establish that IgA alone protects against H. felis infection of the gastric mucosa.
It is also observed that oral administration of H. felis antigen results in significantly increased levels of anti-H. felis IgG antibodies as well as IgA antibodies. There are a number of possible explanations for this phenomenon. First, it has been observed that cholera toxin can, in some cases, enhance both antigen-specific IgA and IgG responses 22. Secondly, cell traffic studies have shown that mesenteric node IgG lymphocytes are a component of the mucosal immune system and can give rise to mucosal IgG plasma cells which have been observed in gastric mucosa. Thirdly, at least a portion of the observed gastric IgG could be the
SUBSTITUTESHEET result of transudation of serum antibody into the gastric lumen secondary to mild to moderate inflammation observed in both control and immunized animals.
The above discussion has focussed on the use of H. felis antigen in the treatment of H. felis infection. It will be appreciated however that the present invention is not limited to the treatment of H. felis infection.
Thus, the present invention also includes within its scope the treatment or prophylaxis of mammals, including humans, for H. pylori infection, wherein the patient is orally immunized with an immunologically effective amount of H. pylori antigen in order to elicit the formation of protective antibodies to H. pylori pathogen. Preferably, the H. pylori is administered in association with a mucosal adjuvant, for example cholera toxin.
Moreover, the present invention includes within its scope the passive immunization of mammals, including humans, against H. pylori infection. This is achieved by orally administering an effective amount of an H. pylori specific antibody to the patient. Preferably an H. pylori specific IgA monoclonal antibody is orally administered to the patient.
The vaccine of the invention is administered orally in amounts readily determined by persons of ordinary skill in this art. Thus, for adults, a suitable dosage would be in the range of lOμg to 10 mg, for example 50μg to 5 mg. Similar dosage ranges would be applicable for children.
SUBSTITUTE SHEET As noted above, a suitable mucosal adjuvant is cholera toxin. Others which may be used are non-toxic derivatives of cholera toxin, including its B subunit and/or conjugates of antigen plus cholera toxin or its B subunit, microcapsules, or immune stimulating complexes (ISCOM's) or liposomes and attenuated live vectors such as viruses or Salmonella bacteria. The amount of mucosal adjuvant employed depends on the type of mucosal adjuvant used. For example, when the mucosal adjuvant is cholera toxin, it is suitably used in an amount of 5μg to 50μg, for example lOμg to 35μg. When used in the form of microcapsules, the amount used will depend on the amount employed in the matrix of the microcapsule to achieve the desired dosage. This is something within the skill of a person of ordinary skill in this art.
Suitable carriers and diluents are enteric coated capsules and/or 0.2N NaHCO., and/or saline. EXAMPLES
The invention will now be further described by the following non-limiting examples. fa) The Mice
The mice used in the experiments were germ-free Swiss Webster mice (8 weeks old) were obtained from Taconic (Germantown, N.Y. ). The animals were housed in microisolater cages under germ-free conditions and they were allowed free access to autoclaved laboratory chows and water. With the exception of occasionally isolating diphtheroidε, animals were maintained in a germ-free state throughout the immunization protocol..
SUBSTITUTE SHEET (b) Bacterial Strains
Bacteria recovered from gastric biopsy specimens of a cat were identified as H. felis based on morphology, Gram stain, and the production of urease, catalase and oxidase . Organisms were stored in 50% phosphate-buffered saline (PBS). 25% glycerol: 25% heated fetal calf serum at -70°C. Bacteria used in the the following examples were passaged in vitro two to three times after isolation.
(c) Bacterial Antigens
The test strain was inoculated onto Columbia agar (Difco, Detroit, MI) containing 7% horse blood and incubated microaerophilically at 37°C for 5-7 days. The organisms were harvested in PBS and the resulting suspensions were sonicated to lyse the bacteria at 40°C, cleared of cellular debris by low-speed centrifugation, and sterile filtered. These whole-cell sonicates were stored as lOOμl aliguots at -70°C until needed for oral immunization of animals.
(d) Outer membranes Outer membranes were prepared as described 19
Briefly, bacterial suspensions were treated with 1 mg of ribonuclease and deoxyribonuclease (Sigma
Chemical, St. Louis) in 0.5 M Tris-EDTA buffer (pH
7.8) at 4°C immediately prior to sonication and low-speed centrifugation as above. Bacterial envelopes were then separated from the cleared lysate by ultracentrifugation at 150,000 x g for 1 h. Outer membranes were separated from the cell envelopes by differential solubilization in sodium n-lauroylsarcosine and recovered by ultracentrifugation. The resulting pellets were
SUBSTITUTE SHEET suspended in 0.05 M phosphate buffer (pH 7.0), divided into aliquots, and stored at -70°C. Protein concentration was determined by the method of Lowry et al ffoorr uussee :in ELISA 20 EXAMPLE 1
Mice were lightly anesthesized by i.p. injection of 1.0 mg ketamine prior to intragastric immunization. Then, whole cell sonicate preparations plus 10 μg of cholera toxin (List Biologicals, Campbell, CA) were suspended in 0.2 M NaHCO,, and 0.5 ml was delivered to the stomachs of mice by intubation through polyethylene tubing attached to a hypodermic syringe. This procedure will be referred to as oral immunization.
To examine the possibility of developing functional immunity, three oral immunization protocols were evaluated. Protocol 1 consisted of 4 oral immunizations over 1 month consisting of 2 mg H. felis lysate plus cholera toxin (a known mucosal adjuvant) . Protocol 2 increased the H. felis to 4 mg per immunization plus cholera toxin, and protocol 3 consisted of 5 oral immunizations over 6 weeks each containing 4 mg of H. felis lysate plus cholera toxin. Unless otherwise noted, animals were challenged 7-10 days after the last immunization and sacrificed 3-7 days later.
The following tissue fluids were collected: serum, gastric secretions, and intestinal secretions. These samples were then titrated for the presence of anti-H. pylori antibodies by enzyme-linked immunosorbent assay (ELISA) . In addition, gastric biopsies were obtained for rapid urease test and culture. Infection was defined as positive if either
SUBSTITUTE SHEET culture or rapid urease test (see below) was positive. Serum was obtained by tail vein bleeding and letting the blood clot at room temperature.
Gastric and intestinal secretions were collected by a modification of the procedure of Elson et al 21'22
Briefly, gastric and intestinal secretions from mice were collected separately. Stomachs and intestines were removed and injected with 2.0 ml of a polyethylene glycol-based lavage plus anti-protease solution. The gastric lavage contained Tris buffer to neutralize gastic acid.
The ELISA was carried out as follows. Murine samples were assayed for H. felis antibodies as follows. Ninety-six well polystyrene microtiter plates were coated with 100 μl/well of appropriate outer membrane proteins (20 μg/ml) overnight at
4°C. Non-specific binding sites were blocked with 1%
BSA in PBS for 90 minutes at room temperature and then the plates were washed with 0.1% BSA in PBS.
Samples were tested in duplicate at dilutions ranging from neat to 1:512,000 and 100 μl of each dilution per well was added to the antigen-coated plates.
Following incubation at room temperature for 90 minutes, the plates were washed three times with 0.1%
BSA in PBS, and 100 μl of a 1:1000 dilution of goat anti-mouse IgA or IgG alkaline phosphataεe conjugate
(Zymed, San Francisco, CA) was added to each well for
90 minutes. After washing, the plates were developed with 100 μl per well of a 1 mg/ml solution of p-nitrophenyl phosphate in glycine buffer (pH 9.6) for 1 hour. The absorbance at 410 nm was measured in each well using a Dynatech MR 700 Microtiter Plate
Reader. The antibody titer was defined as the
SUBSTITUTESHEET reciprocal of the highest dilution yielding an optical density of 0.05 above wells which contained antigen and which were incubated with the antibody conjugate but without the primary antibody sample 18
The rapid urease test was carried out as follows. Two gastric biopsy specimens of 10 mg wet weight from each mouse were immediately placed in 0.2 iriL Stuart urease test broth 28 and incubated at room temperature. The presence of urease was determined by color change from yellow to pink in the test broth after 4 hours 24
Cultures were obtained as follows. Gastric antral biopsies were homogenized and plated onto Columbia agar containing 5% sheep blood, and incubated at 37°C under microaerophilic conditions (gas generating kit; Oxoid Ltd. , London, UK) . A positive culture was defined as visible growth after 5 days. All isolates were identified as H. felis based on morphology, gram stain and the production of urease, catalase and oxidase.
Despite minor changes in experimental design among the three groups, no appreciable differences in immune response were noted. Thus, the data were pooled and the geometric means of gastric lavage, intestinal lavage, and serum antibody titers from the 13 control and 12 immunized animals studied are set forth in Table 1 and Figure 1 discussed above.
Although these animals were both immunized and challenged, the antibody titers did not differ significantly from mice which were immunized and not challenged. In these experiments, gastric, intestinal and serum IgA and IgG antibody titers were significantly higher than that observed in the
SUBSTITUTE SHEET unimmunized control animals. Specifically, there was a 4-fold increase in gastric IgA (p=.001), an 8-fold increase in intestinal IgA (p=.0038) and a 350-fold increase in serum IgA (p=.0001) compared with unimmunized control animals. Similarly, a significant elevation of gastric IgG (p=.0009), intestinal IgG (p=.0001), and serum IgG (p=.0001) was observed.
To evaluate protection from H. felis infection, gastric biopsies were taken from all animals at sacrifice and evaluated by both rapid urease test and culture, as described above. In addition, to determine whether control animals developed a chronic infection and whether immunized animals were definitely H. felis negative, additional immunized and control animals were challenged as above but were not sacrificed until 4 weeks after challenge. The rate of protection among all immunized groups of animals was not appreciably different.
In order to not exclude possible low-level infection, scoring of the gastric biopsy specimens as positive or negative for H. felis growth was not done until 5 days after plating. From plating serial dilutions of known numbers (by hemacytometer count) of culture grown H. felis, it was observed that the sensitivity of this endpoint is approximately 10 organisms. In later experiments, biopsy culture plates were sometimes kept even longer than 5 days and when plates which remained negative for visible growth were scraped and examined by wet mount, an isolated spiral shaped organism could occasionally be seen. The identity of these isolated organisms could not be confirmed, and it could not be determined if
SUBSTITUTESHEET they were viable. In any case, based on the culture results for serially diluted H. felis, it is believed that biopsy εpeciments which remained negative for visible growth at 5 days contained 10 or fewer bacteria.
EXAMPLE 2
IgA and IgA monoclonal antibodies specific for H. felis were produced by a modification of the procedure of Mazanec et al . BALB/c mice obtained from the Jackson Laboratory (Bar Harbor, Maine) were immunized intragastrically four times over a 6-week period, the first three times with 2 mg of sonicated H. felis plus 10 μg of cholera toxin (Sigma Chemical Co., St. Louis, MO). For the last immunization, cholera toxin was omitted, and the mice also received an intravenous boost with 2 mg of H. felis protein. Three days later, the mice were sacrificed, and their spleen cells were hybridized to SP2/0 myeloma cells. Clones, obtained by limiting dilution, were screened for secretion of anti-H. felis IgA antibody by an enzyme-linked immunosorbent assay (ELISA) . The resulting cell line, identified as #71-G5-Aβ, was found to be a stable IgA secreting hybridoma. After multiple Subclonings, stable IgA and IgG secretors were injected intraperitoneally into pristane-primed BALB/c mice, and the ascitic fluid was harvested and clarified.
The cell line #71-G5-Ag, as of April 13, 1992, is deposited in and maintained in viable condition in the Laboratory of Steven J. Czinn, M.D., Rainbow Babies and Children's Hospital, Room 465, Case Western University, 2074 Abington Road, Cleveland, Ohio, U.S.A. 44106. Access to the deposit
SUBSTITUTE SHEET will be available to a person determined by the U.S. Commissioner of Patents and Trademarks to be entitled thereto during pendency of the present application, and all restrictions on availability of the deposit to the public will be irrevocably removed upon grant of a patent on the application.
The cell line #71-G5-Ag is being deposited in the American Type Culture Collection, located at 12301 Parklawn Drive, Rockville, Maryland 20852, U.S.A., under the identification number #71-Gς-Ag. The ATCC accession number and deposit date are , respectively.
EXAMPLE 3
Passive immunization studies were carried out as follows. Ascites containing IgA monoclonal antibody produced from #71-G5-Aβ (200μl) was administered intragastrically simultaneously with 10 viable organisms. Preliminary studies indicated that gastric IgA titers of animals which received a single 200 μl dose of monoclonal IgA antibody declined to levels below that seen in actively immunized animals by 8 hours. Therefore, three additional doses of MAb were given over the next 24 hours. Control animals were challenged identically but received either saline or Sendai virus specific IgA monoclonal antibody (an irrelevant IgA monoclonal antibody) . One week later, the mice were sacrificed. Gastric tissue was inoculated on Columbia blood agar plates and incubated for 5 days at 37°C. Infection was defined as a positive culture or a positive Stuart's rapid urease broth test.
To investigate whether IgA antibodies, the hallmark of the mucosal immune system, could by
SUBSTITUTESHEET themselves protect against H. felis infection of the gastric mucosa, H. felis IgA monoclonal antibodies were generated as described above. One of these antibodies (#71-G5-Ag) was then passively orally administered to germ-free mice at the time of and after challenge with H. felis. Control animals received either saline or Sendai virus specific IgA monoclonal antibody specific for the hemagglutinin-neuraminidase glycoprotein of Sendai virus
The results are presented in Table 2.
TABLE 2
Evaluation of Passive Administration of Antibody To Germ-Free Mice Before and After Challen e with H. felis
Antibody Administered
None-Control
Irrelevant IgA Monoclonal
IgA anti-H. felis monoclonal
H. felis or Sendai virus specific IgA monoclonal antibody were given intragastrically 4 times over 24 hours concurrent with challenge with 10 viable H. felis. Gastric biopsies were obtained 1 week after challenge and infection was determined by culture and/or rapid urease test.
Of the 13 control animals receiving no antibody or Sendai virus antibody, 70% were infected (Figure
SUBSTITUTE SHEET 2B) . Of the seven experimental animals, six were protected and only 1 (14%) was infected. By Chi Square analysis, the difference was significant (p=.019).
Comparison of antibody titers among experimental groups was evaluated by analysis of variance and Fisher's protected T test. For protection, absence or presence of experimental infection among groups were evaluated by Chi Square analysis.
SUBSTITUTE SHEET REFERENCES
1. Blaser, M.J. "Gastric Campylobacter-like organismε, gaεtritiε and peptic ulcer diεeaεe" Gaεtroenterology 1987, 93, 371-183.
2. Graham, D.Y. "Camplyobacter pylori and peptic ulcer disease" Gastroenterology 1989, 96, 615-625.
3. Parsonnet, J. , Vandersteen, D., Goates, J. , Silbey, R.K., Pritkink, J. and Chang, Y. "Helicobacter pylori infection in inteεtinal and diffuse- ype gastric adenocarcinomas" J. Natl. Cancer Inεt. 1991, 93, 640-643.
4. Marshall B.J. , Armstrong, J.A. and McGschie, D.B., "Attempt to fulfill Koch's postulate for pyloric Campylobacter" Med. J. Aust. 1985, 142, 436-439.
5. Morris, A. and Nicholson, H. "Ingeεtion of Campylobacter pyloridiε causeε gaεtritiε and raised faεting gaεtric pH" Am. J. Gaεtroenterol. 1987, 82, 192-199.
6. Engεtrand, L., Guεtavεεon, S., Jδrgenεen, A., Schwann, A., and Schayniuε, A. "Inoculation of barrier-born pigε with Helicobacter pylori: a uεeful animal model for gaεtritiε type B." Infect. Immun. 1990, 53, 1763-1768.
7. Fox, J.G. , Cabot, E.B., Taylor, N.S. and Laraway, R. "Gastric colonization by campylobacter pylori subsp. mustelae in ferrets" Infect. Immun. 1988, 56, 2994-2996.
8. Fox, J.G. , Pelayo, C. , Taylor, N.S., Lee, A., Otto, G. , Murphy, C. and Rose, R. "Helicobacter muεtelae-aεεociated gaεtritiε in ferretε: an animal
SUBSTITUTE SHEET model of Helicobacter pylori gastritis in humans" Gastroenterology 1990, 99, 352-361.
9. Lee, A., Fox, J.G., Otto, G. and Murphy J. "A small animal model of human Helicobacter pylori active chronic gastritis" Gastroenterology 1990, 99, 1315-1323.
10. Fox, J.G., Lee, A., Otto, G., Taylor, N.S. and Murphy J.C. "Helicobacter felis gastritis in gnotobiotic rats: an animal model of helicobacter pylori gastritis" Infect. Immun. 1991, 59, 785-791.
11. Eaton, K.A. , Morgan, D.R. and Krakowka, S. "Campylobacter pylori virulence factors in Gnotobiotic piglets" Infect. Immun. 1989, 57, 1119-1125.
12. Peterson, W.L. "Helicobacter pylori and peptic ulcer'disease" N. Engl. J. Med. 1991, 324,
"1043-1048.
13. Rauwε, E.A.J., Langenberg, W., Houthoff, H.J., Zenon, H.C. and Tytgat, G.N.C. "Campylobacter pyloridiε-aεεociated chronic antral gaεtritiε. A proεpective εtudy of itε prevalence and the effectε of antibacterial and antiulcer treatment" Gastroenterology 1988, 94, 33-40.
14. Fubara, E.S. and Freter, H. "Protection against enteric infection by secretory IgA antibodies" J. Immunol. 1973, 111, 395-403.
15. Offit, P.A. and Clark, H.F. "Protection against rotavirus-induced gaεtroenteritiε in a murine model by passively acquired gastrointestinal but not circulating antibodies" J. Virol. 1985, 54, 58-64.
16. Mazanec, M.B., Nedrud, J.G. and Lamm, M.E. "Immunoglobulin A monoclonal antibodies protect against Sendai virus" J. Virol. 1987, 61, 2624-2626.
SUBSTITUTE SHEET 17. Winner, L.I., Mack, J. , Weltzin, R. ,
McKalanos, J.J., Kraehenbuhl, J.P. and Neutra, M.R.,
"New model for analysis of mucosal immunity:
Intestinal secretion of specific monoclonal immunoglobulin A from hybridoma tumors protect against Vibrio cholerae infection" Infect. Immun.
1991, 59, 977-982.
18. Czinn, S.J. and Nedrud, J.G. "Oral Immunization against Helicobacter pylori" Infect. Immun. 1991, 59, 2359-2363.
19. Sawai, T., Hiruma, R., Kawana, N. , Kaneko, M. Taniyasu, F. and Inami, A. "Outer membrane permeation of beta-lactam antibodies in Eschericia col , Proteus mirabilis and Enterobacter cloacae Antimicrob. Agents." Chemother, 1982, 22, 585-592.
20. Lowry, O.H. , Rosebrough, N.J., Farr, A.J. and Randall, R.J. "Protein measurement with the folin phenol reagent" J. Biol. Chem. 1951, 193, 265-275.
21. Elεon, CO., Ealding, W. and Lefkowitz, J. "A lavage technique allowing repeated measurement of IgA antibody in mouse intestinal secretions" J. Immunol. Meth. 1984, 67, 101-108.
22. Nedrud, J.G., Liang, S., Hague, N. and Lamm, M.E. "Combined oral/nasal immunization protect mice from Sendai viruε infection" J. Immunol. 1987, 139, 3484-3492.
23. Stuart, C, Van Stratum, E. and Ruεtigan, R. "Further studies on urease production by Proteuε and related organiεmε" J. Bacteriol. 1945, 49, 437-444.
24. Czinn, S.J. and Carr, H. "Rapid diagnosis of Campylobacter pyloridiε-aεεociated gaεtritiε, J. Pediatr. 1987, 110-569-570.
25. Brandtzaeg, P. "Role of H chain and
SUBSTITUTE SHEET secretory component in receptor-mediated glandular and hepatic transport of immunoglobulinε in man" Scan. J. Immunol. 1985, 22 , 111-146.
26. Brandtzaeg, P., Bjerka, K. , KEtt, K. , Kvale, D., Rognum, T.O., Scott, H. , Sollid, L.M. and Valnes, K. "Production and secretion of immunoglobulins in the gaεtrointeεtinal tract" Ann. Allergy 1987, 59, 21-39.
27. McDermott, M.R. and Bienenstock, J. "Evidence for a common mucosal immunologic system" I. migration of B. immunoblastε into intestinal, respiratory and genital tissueε" J. Immunol. 1979, 122, 1892-1897.
28. Meεtecky, J. "The common mucosal immune syεtem and current εtrategieε for induction of immune reεponses in external secretions" J. Clin. Immunol. 1987, 7, 265-276.
29. McGhee, J.R., Mestecky, J., Dertzbaugh, M.T., Eldridge, J.H., Hirasawa, M. and Kiyono, H. "The mucosal immune syεtem from fundamental concepts to vaccine development" Vaccine 1992, 10, 75-88.
30. Holmgren, J. , Clemens, J., Sack, D.A. and Svennerholm, A.M. "New cholera vaccines" Vaccineε 1989, 7, 94-96.
31. Ogra, P.L., Karzon, D.T., Righthand, F. and Macgillivray, M. "Immunoglobulin reεponse in serum and secretions after immunization with live and inactivated poliovaccine and natural infection" N. Engl. J. Med. 1968, 279, 895-900.
32. Wyatt, J.I., Rathbone, R.J. and Heatley, R.V. "Local immune responεe to gaεtritic campylobacter in non-ulcer dyspepεiε" J. Clin. Path. 1986, 39, 863-870.
SUBSTITUTE SHEET 33. Kazi, J.I., Sinniah, R. , Jaffrey, N.A., Alam S.M., Zaman, V., Zuberi, S.J. and Kazi, A.M. "Cellular and humoral immune responεe in campylobacter pylori-associated chronic gastritis" J. Pathol. 1989, 159, 231-237.
SUBSTITUTESHEET

Claims (19)

WHAT IS CLAIMED IS:
1. A method of eliciting in a mammalian host a protective immune response to Helicobacter infection, comprising orally adminiεtering to the host an immunogenically effective amount of Helicobacter antigen to elicit said protective human response.
2. A method according to claim 1, wherein said Helicobacter antigen is H. pylori antigen.
3. A method according to claim 1, wherein said Helicobacter antigen is H. felis antigen.
4. A method according to claim 1, wherein said Helicobacter antigen is administered in association with a mucosal adjuvant.
5. A method according to claim 4, wherein said mucosal adjuvant is cholera toxin.
6. A method according to claim 1, wherein said mammalian host is human.
7. A vaccine composition suitable for the treatment of Helicobacter infection, comprising an immunogenically effective amount of Helicobacter antigen for eliciting a protective immune responεe in a mammalian host, in association with a pharmaceutically acceptable carrier or diluent.
8. A vaccine composition according to claim 7, and further comprising an effective amount of a mucosal adjuvant.
9. A vaccine composition according to claim 8, wherein said mucosal adiuvant is cholera toxin.
SUBSTITUTE SHEET
10. A vaccine according to claim 7, wherein said Helicobacter antigen is H. pylori antigen.
11. A vaccine according to claim 7, wherein said Helicobacter antigen is H. felis antigen.
12_ A method of imparting to a mammalian host passive protection to Helicobacter infection, comprising orally administering to εaid host a immunologically effective amount of a Helicobacter specific IgA antibody to impart said passive protection to said host.
13. A method according to claim 12, wherein said antibody is a murine H. felis specific IgA antibody.
14. A method according to claim 13, wherein said antibody is a murine H. felis specific IgA monoclonal antibody produced by cell line #71-G5~Ag.
15. A method according to claim 12, wherein said mammalian host is human.
16. A murine H. felis specific IgA monoclonal antibody.
17. A murine H. felis specific IgA monoclonal antibody.
18. The cell line #71-G5-Ag.
19. A monoclonal antibody produced by cell line
#71-G5-Ag.
SUBSTITUTE SHEET
AU39770/93A 1992-04-13 1993-04-09 Oral treatment of helicobacter infection Withdrawn - After Issue AU675938C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US86828692A 1992-04-13 1992-04-13
US868286 1992-04-13
PCT/US1993/003409 WO1993020843A1 (en) 1992-04-13 1993-04-09 Oral treatment of helicobacter infection

Publications (3)

Publication Number Publication Date
AU3977093A AU3977093A (en) 1993-11-18
AU675938B2 AU675938B2 (en) 1997-02-27
AU675938C true AU675938C (en) 1999-12-02

Family

ID=

Similar Documents

Publication Publication Date Title
US5538729A (en) Oral treatment of helicobacter infection
Czinn et al. Protection of germ-free mice from infection by Helicobacter felis after active oral or passive IgA immunization
US6419926B2 (en) Specific antibodies for use in preparation of pharmaceutical compositions useful in the prevention or treatment of gastritis, gastric ulcers and duodenal ulcers
FI112032B (en) Process for the preparation of a urease-based vaccine against Helicobacter infection
OA10372A (en) Urease-based vaccine and treatment for helicobacter infection
EP0771214B1 (en) Treatment and prevention of helicobacter infection
EP0804489B1 (en) Antigenic preparation for treatment or prevention of helicobacter infection
Zanin et al. Antibody-producing cells in peripheral blood and tonsils after oral treatment of children with bacterial ribosomes
JP2002513812A (en) H. Immunization and treatment against infection with Pylori
AU675938C (en) Oral treatment of helicobacter infection
Vyas et al. Exploring novel vaccines against Helicobacter pylori: protective and therapeutic immunization