Helicobacter pylori

Listen to this article
Source: Wikipedia, the free encyclopedia.

Helicobacter pylori
negative staining
)
Scientific classification Edit this classification
Domain: Bacteria
Phylum: Campylobacterota
Class:
"Campylobacteria"
Order: Campylobacterales
Family:
Helicobacteraceae
Genus: Helicobacter
Species:
H. pylori
Binomial name
Helicobacter pylori
(Marshall et al. 1985) Goodwin et al., 1989
Synonyms
  • Campylobacter pylori Marshall et al. 1985

Helicobacter pylori, previously known as Campylobacter pylori, is a

gastric ulcers in 1983 by the Australian doctors Barry Marshall and Robin Warren.[4][5]

Infection of the stomach with H. pylori is not the cause of illness itself; over half of the global population is infected but most are asymptomatic.

gastric cancer.[9][10] Infection with H. pylori is responsible for around 89 per cent of all gastric cancers, and is linked to the development of 5.5 per cent of all cases of cancer worldwide.[12][13] H. pylori is the only bacterium known to cause cancer.[14]

Extragastric complications that have been linked to H. pylori include

GERD and Crohn's disease) and other disorders.[15]

Some studies suggest that H. pylori plays an important role in the natural stomach ecology by influencing the type of bacteria that colonize the gastrointestinal tract.[16][17] Other studies suggest that non-pathogenic strains of H. pylori may beneficially normalize stomach acid secretion, and regulate appetite.[18]

In 2023, it was estimated that about two-thirds of the world's population were infected with H. pylori, being more common in

standards of living.[20][21]

Microbiology

Helicobacter pylori is a species of gram-negative bacteria in the Helicobacter genus.[22] About half the world's population is infected with H. pylori but only a few strains are

cell wall's peptidoglycan.[1] The bacteria reach the less acidic mucosa by use of their flagella.[25] Three strains studied showed a variation in length from 2.8–3.3 μm but a fairly constant diameter of 0.55–0.58 μm.[23] H. pylori can convert from a helical to an inactive coccoid form, that may possibly become viable, known as viable but nonculturable (VBNC).[26]

Helicobacter pylori is

atmosphere. It contains a hydrogenase that can produce energy by oxidizing molecular hydrogen (H2) made by intestinal bacteria.[27]

H. pylori can be demonstrated in tissue by Gram stain, Giemsa stain, H&E stain, Warthin-Starry silver stain, acridine orange stain, and phase-contrast microscopy. It is capable of forming biofilms. Biofilms help to hinder the action of antibiotics and can contribute to treatment failure.[28][29]

To successfully colonize H. pylori uses many different virulence factors including oxidase, catalase, and urease.[30] Urease is the most abundant protein, its expression representing about 10% of the total protein weight.[31]

H. pylori possesses five major

phospholipids and lipopolysaccharide (LPS). The O-antigen of LPS may be fucosylated and mimic Lewis blood group antigens found on the gastric epithelium.[30]

Genome

Helicobacter pylori consists of a large diversity of strains, and hundreds of genomes have been completely sequenced.[32][33][34] The genome of the strain 26695 consists of about 1.7 million base pairs, with some 1,576 genes.[35][36] The pan-genome, that is the combined set of 30 sequenced strains, encodes 2,239 protein families (orthologous groups OGs).[37] Among them, 1,248 OGs are conserved in all the 30 strains, and represent the universal core. The remaining 991 OGs correspond to the accessory genome in which 277 OGs are unique to one strain.[38]

There is an unusually high number of restriction modification systems in the genome of H. pylori.[39] These provide a defence against bacteriophages.[39]

Transcriptome

Shine–Dalgarno sequence in H. pylori.[40]

Proteome

The proteome of H. pylori has been systematically analyzed and more than 70% of its proteins have been detected by mass spectrometry, and other methods. About 50% of the proteome has been quantified, informing of the number of protein copies in a typical cell.[41]

Studies of the

protein-protein interactions. This has provided information of how proteins interact with each other, either in stable protein complexes or in more dynamic, transient interactions, which can help to identify the functions of the protein. This in turn helps researchers to find out what the function of uncharacterized proteins is, e.g. when an uncharacterized protein interacts with several proteins of the ribosome (that is, it is likely also involved in ribosome function). About a third of all ~1,500 proteins in H. pylori remain uncharacterized and their function is largely unknown.[42]

Infection

Diagram of stages of ulcer development

An infection with Helicobacter pylori can either have no symptoms even when lasting a lifetime, or can harm the stomach and duodenal

gastric cancer, known as Correa's cascade.[46][47] Extragastric complications that have been linked to H. pylori include anemia due either to iron-deficiency or vitamin B12 deficiency, diabetes mellitus, cardiovascular, and certain neurological disorders.[15]

Peptic ulcers are a consequence of inflammation that allows stomach acid and the digestive enzyme

mucous membranes. The location of colonization of H. pylori, which affects the location of the ulcer, depends on the acidity of the stomach.[48]
In people producing large amounts of acid, H. pylori colonizes near the
fundus (near the entrance to the stomach).[30] G cells express relatively high levels of PD-L1 that protects these cells from H. pylori-induced immune destruction.[49]
In people producing normal or reduced amounts of acid, H. pylori can also colonize the rest of the stomach.

Diagram showing parts of the stomach

The inflammatory response caused by bacteria colonizing near the pyloric antrum induces G cells in the antrum to secrete the hormone gastrin, which travels through the bloodstream to parietal cells in the fundus.[50] Gastrin stimulates the parietal cells to secrete more acid into the stomach lumen, and over time increases the number of parietal cells, as well.[51] The increased acid load damages the duodenum, which may eventually lead to the formation of ulcers.

Helicobacter pylori is a class I

gastric cancer.[9][10][52] Less commonly diffuse large B-cell lymphoma of the stomach is a risk.[53] Infection with H. pylori is responsible for around 89 per cent of all gastric cancers, and is linked to the development of 5.5 per cent of all cases of cancer worldwide.[12][13] Although the data varies between different countries, overall about 1% to 3% of people infected with Helicobacter pylori develop gastric cancer in their lifetime compared to 0.13% of individuals who have had no H. pylori infection.[54][30] H. pylori-induced gastric cancer is the third highest cause of worldwide cancer mortality as of 2018.[55] Because of the usual lack of symptoms, when gastric cancer is finally diagnosed it is often fairly advanced. More than half of gastric cancer patients have lymph node metastasis when they are initially diagnosed.[56]

Micrograph of H. pylori colonizing the stomach lining

Chronic inflammation that is a feature of cancer development is characterized by infiltration of

double-strand breaks.[59]

Small

gastric and colorectal polyps are adenomas that are more commonly found in association with the mucosal damage induced by H. pylori gastritis.[60][61] Larger polyps can in time become cancerous.[62][60] A modest association of H. pylori has been made with the development of colorectal cancers but as of 2020 causality had yet to be proved.[63][62]

Signs and symptoms

Most people infected with H. pylori never experience any symptoms or complications, but will have a 10% to 20% risk of developing

belching, feeling hunger in the morning, feeling full too soon, and sometimes vomiting, heartburn, bad breath, and weight loss.[66][67]

Complications of an ulcer can cause severe signs and symptoms such as black or tarry stool indicative of

corpus
may lead to a gastric ulcer.

Gastric polyps are adenomas that are usually asymptomatic and benign, but may be the cause of dyspepsia, heartburn, bleeding from the stomach, and, rarely, gastric outlet obstruction.[60][70] Larger polyps may have become cancerous.[60]
Colorectal polyps may be the cause of rectal bleeding, anemia, constipation, diarrhea, weight loss, and abdominal pain.[71]

Pathophysiology

Virulence factors help a pathogen to evade the immune response of the host, and to successfully colonize. The many virulence factors of H. pylori include its flagella, the production of urease, adhesins, serine protease HtrA (high temperature requirement A), and the major exotoxins CagA and VacA.[28][72] The presence of VacA and CagA are associated with more advanced outcomes.[73] CagA is an oncoprotein associated with the development of gastric cancer.[6]

Diagram of H. pylori and associated virulence factors
Diagram showing how H. pylori reaches the epithelium of the stomach

H. pylori infection is associated with

MGMT and MRE11 are also evident. Reduced DNA repair in the presence of increased DNA damage increases carcinogenic mutations and is likely a significant cause of gastric carcinogenesis.[58][77][78] These epigenetic alterations are due to H. pylori-induced methylation of CpG sites in promoters of genes[77] and H. pylori-induced altered expression of multiple microRNAs.[78]

Two related mechanisms by which H. pylori could promote cancer have been proposed. One mechanism involves the enhanced production of

free radicals near H. pylori and an increased rate of host cell mutation. The other proposed mechanism has been called a "perigenetic pathway",[79] and involves enhancement of the transformed host cell phenotype by means of alterations in cell proteins, such as adhesion proteins. H. pylori has been proposed to induce inflammation and locally high levels of tumor necrosis factor (TNF), (also known as tumor necrosis factor alpha (TNFα) and/or interleukin 6 (IL-6). According to the proposed perigenetic mechanism, inflammation-associated signaling molecules, such as TNF, can alter gastric epithelial cell adhesion and lead to the dispersion and migration of mutated epithelial cells without the need for additional mutations in tumor suppressor genes, such as genes that code for cell adhesion proteins.[80]

Flagellum

The first virulence factor of Helicobacter pylori that enables colonization is its

the same polar location which gives it a high motility. The flagellar filaments are about 3 μm long, and composed of two copolymerized flagellins, FlaA and FlaB, coded by the genes flaA, and flaB.[25][72] The minor flagellin FlaB is located in the proximal region and the major flagellin FlaA makes up the rest of the flagellum.[82] The flagella are sheathed in a continuation of the bacterial outer membrane which gives protection against the gastric acidity. The sheath is also the location of the origin of the outer membrane vesicles that gives protection to the bacterium from bacteriophages.[82]

H. pylori is able to sense the less acidic

gastric glands.[83] Occasionally the bacteria are found inside the epithelial cells themselves.[84]

Urease

H. pylori urease enzyme diagram

In addition to using chemotaxis to avoid areas of low pH (high acidity), H. pylori also neutralizes the acid in its environment by producing large amounts of urease, an enzyme which breaks down the urea present in the stomach to carbonic acid and ammonia. These react with the strong acids in the environment to produce a neutralized area around H. pylori.[85] Helicobacter pylori is one of the few known types of bacterium that has a urea cycle which is uniquely configured in the bacterium.[86] 10% of the cell is of nitrogen a balance that needs to be maintained. Any excess is stored in urea excreted in the urea cycle.[86]

A final stage enzyme in the urea cycle is arginase an enzyme that is crucial to the pathogenesis of H. pylori. Arginase produces ornithine and urea that the enzyme urease breaks down into carbonic acid and ammonia. Urease is the bacterium’s most abundant protein accounting for 10–15% of the bacterium's total protein content. Its expression is not only required for establishing initial colonization in the breakdown of urea to carbonic acid and ammonia but is essential for maintaining chronic infection.[87][64] Ammonia reduces the stomach acidity allowing the bacteria to become locally established. Arginase promotes the persistence of infection by consuming arginine; arginine is used by macrophages to produce nitric oxide which has a strong antimicrobial effect.[86][88] The ammonia produced to regulate pH is toxic to epithelial cells. [89]

Adhesins

H. pylori must make attachment with the epithelial cells to prevent its being swept away with the constant movement and renewal of the mucus. To give them this adhesion,

sialyl-Lewis X antigen expressed on gastric mucosa.[93]

Cholesterol glucoside

The outer membrane contains cholesterol glucoside, a sterol glucoside that H. pylori glycosylates from the cholesterol in the gastric gland cells, and inserts it into its outer membrane.[85] This cholesterol glucoside is important for membrane stability, morphology and immune evasion, and is rarely found in other bacteria.[94][95]

The enzyme responsible for this is cholesteryl α-glucosyltransferase (αCgT) or (Cgt) encoded by the HP0421 gene.[96] A major effect of the depletion of host cholesterol by Cgt is to disrupt cholesterol-rich lipid rafts in the epithelial cells. Lipid rafts are involved in cell signalling and their disruption causes a reduction in the immune inflammatory response particularly by reducing interferon gamma.[97] Cgt is also secreted by the type IV secretion system, and is secreted in a selective way so that gastric niches where the pathogen can thrive are created.[96] Its lack has been shown to give vulnerability from environmental stress to bacteria, and also to disrupt CagA mediated interactions.[85]

Catalase

Colonization induces an intense anti-inflammatory response as a first-line immune system defence. Phagocytic leukocytes and monocytes infiltrate the site of infection, and antibodies are produced.[98] H. pylori is able to adhere to the surface of the phagocytes and impede their action. This is responded to by the phagocyte in the generation and release of oxygen metabolites into the surrounding space. H. pylori can survive this response by the activity of catalase at its attachment to the phagocytic cell surface. Catalase decomposes hydrogen peroxide into water and oxygen, protecting the bacteria from toxicity. Catalase has been shown to almost completely inhibit the phagocytic oxidative response.[98] It is coded for by the gene katA.[99]

Tipα

TNF-inducing protein alpha (Tipα) is a carcinogenic protein encoded by HP0596 unique to H. pylori that induces the expression of tumor necrosis factor.[80][100] Tipα enters gastric cancer cells where it binds to cell surface nucleolin, and induces the expression of vimentin. Vimentin is important in the epithelial–mesenchymal transition associated with the progression of tumors.[101]

CagA

CagA (cytotoxin-associated antigen A) is a major

oncoprotein that is encoded by the cagA gene. Bacterial strains with the cagA gene are associated with the ability to cause ulcers, MALT lymphomas, and gastric cancer.[102][103] The cagA gene codes for a relatively long (1186-amino acid) protein. The cag pathogenicity island (PAI) has about 30 genes, part of which code for a complex type IV secretion system. The low GC-content of the cag PAI relative to the rest of the Helicobacter genome suggests the island was acquired by horizontal transfer from another bacterial species.[36] The serine protease HtrA also plays a major role in the pathogenesis of H. pylori. The HtrA protein enables the bacterium to transmigrate across the host cells' epithelium, and is also needed for the translocation of CagA.[104]

The virulence of H. pylori may be increased by genes of the cag pathogenicity island; about 50–70% of H. pylori strains in Western countries carry it.

cytokines that promote inflammation.[106]

The type-IV

These proteins are directly toxic to cells lining the stomach and signal strongly to the immune system that an invasion is under way. As a result of the bacterial presence, neutrophils and macrophages set up residence in the tissue to fight the bacteria assault.
gene transcription, independent of protein tyrosine phosphorylation.[103]
A great deal of diversity exists between strains of H. pylori, and the strain that infects a person can predict the outcome.

VacA

VacA (vacuolating cytotoxin auto transporter) is another major virulence factor encoded by the vacA gene.

COX2, an up-regulation that increases the production of a prostaglandin indicating a strong host cell inflammatory response.[112][114]

Outer membrane proteins and vesicles

About 4% of the genome encodes for

phospholipids and lipopolysaccharide (LPS). The O-antigen of LPS may be fucosylated and mimic Lewis blood group antigens found on the gastric epithelium.[30]

H. pylori forms blebs from the outer membrane that pinch off as outer membrane vesicles to provide an alternative delivery system for virulence factors including CagA.[85]

A Helicobacter cysteine-rich protein HcpA is known to trigger an immune response, causing inflammation.[116] A Helicobacter pylori virulence factor DupA is associated with the development of duodenal ulcers.[117]

Mechanisms of tolerance

In the stomach H. pylori has to not only survive the harsh gastric acidity but also the constant sweeping of mucus by continuous

DNA damage is supported by transformation-mediated recombinational repair, that contributes to successful colonization.[120][121] An overall response to multiple stressors can result from an interaction of the mechanisms. The mechanisms of tolerance and persistence can also help to overcome the effects of antibiotics.[119]

An effective sustained colonization response is the formation of a biofilm. Layers of aggregated bacteria form a biofilm. Cells in the deep layers are nutritionally deprived, and enter the coccoid dormant-like state. Some of these cells will be antibiotic resistant, and may remain in the host as persister cells. Following eradication the persister cells can cause a recurrence of the infection.[122][123]

double-strand breaks (DSBs). The AddAB helicase-nuclease complex resects DSBs and loads RecA onto single-strand DNA (ssDNA), which then mediates strand exchange, leading to homologous recombination and repair. The requirement of RecA plus AddAB for efficient gastric colonization suggests, in the stomach, H. pylori is either exposed to double-strand DNA damage that must be repaired or requires some other recombination-mediated event. In particular, natural transformation is increased by DNA damage in H. pylori, and a connection exists between the DNA damage response and DNA uptake in H. pylori,[124] suggesting natural competence contributes to persistence of H. pylori in its human host and explains the retention of competence in most clinical isolates. H. pylori has much greater rates of recombination, and mutation than other bacteria.[3] Genetically different strains can be found in the same host, and also in different regions of the stomach.[125]

RuvC protein is essential to the process of recombinational repair, since it resolves intermediates in this process termed Holliday junctions. H. pylori mutants that are defective in RuvC have increased sensitivity to DNA-damaging agents and to oxidative stress, exhibit reduced survival within macrophages, and are unable to establish successful infection in a mouse model.[126] Similarly, RecN protein plays an important role in DSB repair in H. pylori.[127] An H. pylori recN mutant displays an attenuated ability to colonize mouse stomachs, highlighting the importance of recombinational DNA repair in survival of H. pylori within its host.[127]

Diagnosis

H. pylori colonized on the surface of regenerative epithelium (Warthin-Starry silver stain)

Colonization with H. pylori is not a disease in itself, but a condition associated with a number of

iron deficiency anemia, or in cases of immune thrombocytopenic purpura.[128]
Several methods of testing exist, both invasive and non-invasive.

Non-invasive tests for H. pylori infection include

antibodies, stool tests, and urea breath tests. Carbon urea breath tests include the use of carbon-13, or a radioactive carbon-14 producing a labelled carbon dioxide that can be detected in the breath.[129] Carbon urea breath tests have a high sensitivity and specificity for the diagnosis of H. pylori.[129]

Proton-pump inhibitors and antibiotics should be discontinued for at least 30 days prior to testing for H. pylori infection or eradication, as both agents inhibit H. pylori growth and may lead to false negative results.[128] Testing to confirm eradication is recommended 30 days or more after completion of treatment for H. pylori infection. H. pylori breath testing or stool antigen testing are both reasonable tests to confirm eradication.[128] H. pylori serologic testing, including IgG antibodies, are not recommended as a test of eradication as they may remain elevated for years after successful treatment of infection.[128]

An endoscopic biopsy is an invasive means to test for H. pylori infection. Low-level infections can be missed by biopsy, so multiple samples are recommended. The most accurate method for detecting H. pylori infection is with a

histological examination from two sites after endoscopic biopsy, combined with either a rapid urease test or microbial culture.[130] Generally, repeating endoscopy is not recommended to confirm H. pylori eradication, unless there are specific indications to repeat the procedure.[128]

Transmission

Helicobacter pylori is contagious, and transmission is through either the oral–oral route or the fecal–oral route, but is mainly associated with the oral–oral route.[7] Consistent with these transmission routes, the bacteria have been isolated from feces, saliva, and dental plaque.[131] H. pylori may also be transmitted orally by drinking contaminated water.[7] Transmission occurs mainly within families in developed nations, yet can also be acquired from the community in developing countries.[132]

Prevention

To prevent the development of H. pylori-related diseases when infection is suspected, antibiotic-based therapy regimens are recommended to eradicate the bacteria.[45] When successful the disease progression is halted. First line therapy is recommended if low-grade gastric MALT lymphoma is diagnosed, regardless of evidence of H. pylori. However, if a severe condition of atrophic gastritis with gastric lesions is reached antibiotic-based treatment regimens are not advised since such lesions are often not reversible and will progress to gastric cancer.[45] If the cancer is managed to be treated it is advised that an eradication program be followed to prevent a recurrence of infection, or reduce a recurrence of the cancer, known as metachronous.[45][133][134]

Due to H. pylori's role as a major cause of certain diseases (particularly cancers) and its consistently increasing

resistance to antibiotic therapy, there is an obvious need for alternative treatments.[135] A vaccine targeted towards the development of gastric cancer including MALT lymphoma, would also prevent the development of gastric ulcers.[5] A vaccine that would be prophylactic for use in children, and one that would be therapeutic later are the main goals. Challenges to this are the extreme genomic diversity shown by H. pylori and complex host-immune responses.[135][136]

Previous studies in the Netherlands, and in the US have shown that such a prophylactic vaccine programme would be ultimately cost-effective.[137][138] However, as of late 2019 there have been no advanced vaccine candidates and only one vaccine in a Phase I clinical trial. Furthermore, development of a vaccine against H. pylori has not been a priority of major pharmaceutical companies.[139] A key target for potential therapy is the proton-gated urea channel, since the secretion of urease enables the survival of the bacterium.[140]

Treatment

Gastritis

Following Maastricht Consensus Reports, H. pylori gastritis, has been included in

ICD11, and listed as Helicobacter pylori induced gastritis.[43][44][45] Initially the infection tends to be superficial, localised to the upper mucosal layers of the stomach.[141] The intensity of chronic inflammation is related to the cytotoxicity of the H. pylori strain. A greater cytotoxicity will result in the change from a non-atrophic gastritis to an atrophic gastritis with the loss of mucous glands. This condition is a prequel to the development of peptic ulcers and gastric adenocarcinoma.[141]

Various antibiotic plus proton-pump inhibitor drug regimens are used to eradicate the infection and thereby successfully treat the disorder[142] with triple-drug therapy consisting of clarithromycin, amoxicillin, and a proton-pump inhibitor given for 14–21 days often being considered first line treatment.[141]

Peptic ulcers

Once H. pylori is detected in a person with a peptic ulcer, the normal procedure is to eradicate it and allow the ulcer to heal. The standard

H2-antagonists or proton pump inhibitors alone.[148][149] Eradication of H. pylori is associated with a subsequent decreased risk of duodenal or gastric ulcer recurrence.[128]

Antibiotic resistance

Increasing

Next generation sequencing is looked to for identifying initial specific antibiotic resistances that will help in targeting more effective treatment.[152]

In 2018 the

WHO listed H. pylori as a high priority pathogen for the research and discovery of new drugs and treatments.[153] The increasing antibiotic resistance encountered has spurred interest in developing alternative therapies using a number of plant compounds.[154][155] Plant compounds have fewer side effects than synthetic drugs. Most plant extracts contain a complex mix of components that may not act on their own as antimicrobials but can work together with antibiotics to enhance treatment and work towards overcoming resistance.[154] Plant compounds have a different mechanism of action that has proved useful in fighting antimicrobial resistance. Various compounds can act for example by inhibiting enzymes such as urease, and adhesions to the mucous membrane.[156] Sulfur-containing compounds from plants with high concentrations of polysulfides, coumarins, and terpenes have all been shown to be effective against H. pylori.[154]

Additional rounds of antibiotics may be used or other therapies.[157][158][159] In patients with any previous macrolide exposure or who are allergic to penicillin, a quadruple therapy that consisting of a proton pump inhibitor, bismuth, tetracycline, and a nitroimidazole for 10–14 days is a recommended first-line treatment option.[160] For the treatment of clarithromycin-resistant strains of H. pylori, the use of levofloxacin as part of the therapy has been suggested.[161][162]

butyrate that acts as a prebiotic and enhances the mucosal immune barrier. Their use as probiotics may help balance the gut dysbiosis that accompanies antibiotic use.[163] Some probiotic strains have been shown to have bactericidal and bacteriostatic activity against H. pylori, and also help to balance the gut dysbiosis.[164]

H. pylori is found in saliva and dental plaque. Its transmission is known to include oral-oral suggesting that the dental plaque may act as a reservoir for the bacteria. Periodontal therapy or scaling and root planing has therefore been suggested as an additional treatment to enhance eradication rates but more research is needed.[165]

Cancers

Stomach cancer

Helicobacter pylori is linked to the majority of

gastroesophageal junction.[166] The treatment for this cancer is highly aggressive with even localized disease being treated sequentially with chemotherapy and radiotherapy before surgical resection.[167] Since this cancer, once developed, is independent of H. pylori infection, antibiotic-proton pump inhibitor regimens are not used in its treatment.[166]

Gastric MALT lymphoma and DLBCL

MALT lymphomas are malignancies of mucosa-associated lymphoid tissue. Early gastric MALTomas due to H. pylori may be successfully treated (70–95% of cases) with one or more eradication programs.[13] Some 50–80% of patients who experience eradication of the pathogen develop within 3–28 months a remission and long-term clinical control of their lymphoma. Radiation therapy to the stomach and surrounding (i.e. peri-gastric) lymph nodes has also been used to successfully treat these localized cases. Patients with non-localized (i.e. systemic Ann Arbor stage III and IV) disease who are free of symptoms have been treated with watchful waiting or, if symptomatic, with the immunotherapy drug, rituximab, (given for 4 weeks) combined with the chemotherapy drug, chlorambucil, for 6–12 months; 58% of these patients attain a 58% progression-free survival rate at 5 years. Frail stage III/IV patients have been successfully treated with rituximab or the chemotherapy drug, cyclophosphamide, alone.[168] Antibiotic-proton pump inhibitor eradication therapy and localized radiation therapy have been used successfully to treat H. pylori-positive MALT lymphomas of the rectum; however radiation therapy has given slightly better results and therefore been suggested to be the disease' preferred treatment.[169] However, the generally recognized treatment of choice for patients with systemic involvement uses various chemotherapy drugs often combined with rituximab.

A MALT lymphoma may rarely transform into a more aggressive diffuse large B-cell lymphoma (DLBCL).[170] Where this is associated with H. pylori infection the DLBCL is less aggressive and more amenable to treatment.[171][172][173] When limited to the stomach they have sometimes been successfully treated with H. pylori eradication programs.[53][172][174][173] If unresponsive or showing a deterioration, a more conventional chemotherapy (CHOP), immunotherapy or local radiotherapy can be considered, and any of these or a combination have successfully treated these more advanced types. [172][173]

Prognosis

Helicobacter pylori colonizes the stomach for decades in most people, and induces chronic gastritis, a long-lasting inflammation of the stomach. In most cases symptoms are never experienced but about 10–20% of those infected will ultimately develop gastric and duodenal ulcers, and have a possible 1–2% lifetime risk of gastric cancer.[64]

H. pylori thrives in a high salt diet, which is seen as an environmental risk factor for its association with gastric cancer. A diet high in salt enhances colonization, increases inflammation, increases the expression of H. pylori virulence factors, and intensifies chronic gastritis.[175][176] Paradoxically extracts of kimchi a salted probiotic food has been found to have a preventive effect on H. pylori associated gastric carcinogenesis.[177]

In the absence of treatment, H. pylori infection, usually persists for life.[178] Infection may disappear in the elderly as the stomach's mucosa becomes increasingly atrophic and inhospitable to colonization. Some studies in young children up to two years of age, have shown that infection can be transient in this age group.[179][180]

It is possible for H. pylori to re-establish in a person after eradication. This recurrence can be caused by the original strain (recrudescence), or be caused by a different strain (reinfection). A 2017 meta-analysis showed that the global per-person annual rates of recurrence, reinfection, and recrudescence is 4.3%, 3.1%, and 2.2% respectively. It is unclear what the main risk factors are.[181]

Mounting evidence suggests H. pylori has an important role in protection from some diseases.

gastric microbiota.[16] He postulates that the changes in gastric physiology caused by the loss of H. pylori account for the recent increase in incidence of several diseases, including type 2 diabetes, obesity, and asthma.[16][185] His group has recently shown that H. pylori colonization is associated with a lower incidence of childhood asthma.[186]

Epidemiology

In 2023, it was estimated that about two-thirds of the world's population were infected with H. pylori infection, being more common in

developing countries.[19] H. pylori infection is more prevalent in South America, Sub-Saharan Africa, and the Middle East.[142] The global prevalence declined markedly in the decade following 2010, with a particular reduction in Africa.[20]

The age when someone acquires this bacterium seems to influence the pathologic outcome of the infection. People infected at an early age are likely to develop more intense inflammation that may be followed by atrophic gastritis with a higher subsequent risk of gastric ulcer, gastric cancer, or both. Acquisition at an older age brings different gastric changes more likely to lead to duodenal ulcer.

African-American and Hispanic populations, most likely due to socioeconomic factors.[187][188] The lower rate of infection in the West is largely attributed to higher hygiene standards and widespread use of antibiotics. Despite high rates of infection in certain areas of the world, the overall frequency of H. pylori infection is declining.[189] However, antibiotic resistance is appearing in H. pylori; many metronidazole- and clarithromycin-resistant strains are found in most parts of the world.[190]

History