Helicobacter pylori cagA Status and Gastric Mucosa-associated Lymphoid Tissue Lymphoma: a Systematic Review and Meta-analysis

DOI: https://doi.org/10.21203/rs.3.rs-289927/v1

Abstract

Background: Recent studies have investigated the role of Helicobacter pylori infection in the development of gastric mucosa-associated lymphoid tissue (MALT) lymphoma. It is estimated that approximately 0.1% of people infected with H. pylori develop gastric MALT. However, the role of the CagA antigen, the highest causative agent of H. pylori, in increasing the risk of gastric MALT remains unclear and controversial. A systematic review and meta-analysis were conducted to evaluate the effect of cagA status on the development of gastric MALT.

Methods: All articles evaluating the status of the cagA gene in the development of gastric MALT were collected using systematic searches in online databases, including PubMed, Scopus, Embase, and Google Scholar, regardless of publication date. The association between cagA and gastric MALT was assessed using the odds ratio (OR) summary. In addition, a random-effects model was used in cases with significant heterogeneity.

Results: A total of 10 studies met our inclusion criteria, among which 1,860 patients participated. We observed a meaningful association between cagA status and gastric MALT, especially in Western countries (OR: 1.30; 0.90–1.87 with 95% CIs). However, the heterogeneity was significant; therefore, the results should be interpreted with caution. Surprisingly, a strong positive association was observed between cagA status and high-grade lymphomas (OR: 6.43; 2.45–16.84 with 95% CIs). In addition, no study has evaluated the correlation between cagA and vacA, but we observed an inverse association between vacA and gastric MALT risk (OR: 0.92; 0.57–1.50 with 95% CIs).

Conclusion: CagA may not play a significant role in the early stages of gastric MALT, but it can be translocated to B cells and affect the development of high-grade lymphomas.

Introduction

Helicobacter pylori (H. pylori) is one of the most unique human pathogenic bacteria, and because of its exceptional ability to tolerate harsh stomach conditions, it colonizes the stomachs of about 4–4.5 billion people worldwide[1, 2]. Depending on environmental, socioeconomic, and health conditions, the prevalence of H. pylori infection varies in different geographical areas, with values of about 34% in Western countries and close to 100% in developing countries[3, 4]. Bacterial strains enter the gastric submucosa and cause chronic gastritis by evading the immune system response. However, 15–20% of people infected with H. pylori experience severe clinical outcomes, especially peptic ulcer disease (gastric ulcer or duodenal ulcer), chronic atrophy gastritis, and gastric adenocarcinoma, e.g., gastric cancer or mucosa-associated lymphoid tissue (MALT) lymphoma[5, 6].

It is currently unknown why most people infected with H. pylori are asymptomatic carriers, and severe clinical outcomes are seen only in a small part of the human population. The genomic content of H. pylori is specific to the strain, and evaluating the role of strain virulence factors is critical[7, 8]. The cytotoxin-associated gene A (CagA) is one of the major virulence factors in this bacterium, which is encoded by the cag pathogenicity islands (PAIs) and is classified into four different classes based on the flanking nucleotide sequence of EPIYA motifs. The CagA pattern in the East Asian population is usually ABD, while strains containing patterns such as ABC, ABCC, and ABCCC are isolated from Western countries[9, 10]. According to previous studies, cagA-positive strains are significantly present in the population with gastric ulcers and precancerous lesions[11]. Although the role of CagA in tumorigenesis remains unclear, according to the first hypothesis, phosphorylated CagA can phosphorylate intracellular eukaryotic proteins, particularly SHP-2 and Src kinase, and induce the hummingbird phenotype and oncogenesis by altering the normal cell signaling pathway[12]. The affinity of EPIYA-D for the binding and effect of SHP2 tyrosine phosphatase is much higher than that of EPIYA-C, so the differences are reported to be related to the fact that the prevalence of gastric cancer in East Asia is higher than that in Western countries[11, 13].

According to the second hypothesis, CagA is an immunogenic protein that stimulates the ‎production of interleukin (IL)-8 and leads to the infiltration of neutrophils in the inflamed area, the production of free ‎radicals, and DNA damage[1416]. Accordingly, infection with this bacterium appears to increase the risk of two cancers of the digestive system, including gastric cancer and gastric MALT[17].

MALT lymphoma was first identified by Isaacson and Wright in 1983 and accounts for more than 50% of gastric lymphoma, a B-cell lymphoma derived from MALT during chronic inflammation[17, 18]. Carlson et al. (1996) showed that H. pylori gastritis can lead to gastric MALT by causing polyclonal lymphatic hyperplasia[19]. Recently, it has been shown that there is a positive association between H. pylori infection and the development of gastric MALT, with more than 90% of patients with gastric MALT lymphoma being infected with H. pylori[20]. In a study by Asenjo et al., the global prevalence of H. pylori infection in high-grade and low-grade lymphomas was estimated to be about 60% and 79%, respectively[21]. Interestingly, the eradication of H. pylori infection is very effective in the regression of gastric MALT; therefore, antibiotic therapy is considered the first line of treatment for gastric MALT[22]. According to in vitro experiments, the immune response in low-grade lymphomas is formed as a result of T cell-mediated immunity[23, 24]. Studies have shown that the CagA antigen can be translocated to B-cell lymphocytes following the destruction of gastric mucosa during chronic gastritis[25, 26]. In B cells, this antigen prevents apoptosis through extracellular signal-regulated kinase, which in turn leads to the proliferation and immortalization of B cells and eventually MALT lymphoma[2628].

In general, despite limited information and conflicting results in some studies, events such as chronic inflammation, production of reactive oxygen species (ROS), B-cell proliferation, and genetic instability can lead to susceptibility to gastric MALT lymphoma[2931]. In the present meta-analysis, we investigated the association between CagA and MALT to evaluate the role of CagA in the development of gastric MALT lymphoma.

Materials And Methods

Literature search

We conducted a comprehensive electronic search using the following online databases: PubMed, Scopus, Embase, and Google Scholar to retrieve all relevant documents printed in English up until December 2020. The search for terms was performed based on the MeSH library using words such as “Helicobacter pylori,” “H. pylori,” "MALT,” “mucosa-associated lymphoid tissue,” “CagA,” and “cytotoxin-associated gene A.” The literature search was performed independently by two authors (MK1 and MK2) without publication date restrictions.

Study selection

In the first stage, after initial evaluations, duplicate articles were excluded from the study, and then a reference list of each article was evaluated to avoid losing additional documents. The inclusion criteria were as follows: 1) all original, cross-sectional, case-control, and longitudinal articles related to our purpose, 2) studies on the association between cagA gene status and gastric MALT lymphoma; 3) studies based on standard diagnostic methods such as polymerase chain reaction (PCR), ELISA, and conventional microbiology tests; and 4) studies published in English. Exclusion criteria were as follows: (i) congress abstracts, case series, review articles, and letters to the editor; II) articles without full text available; III) articles published in non-English language; IV) animal studies or in vitro studies; and V) studies with vague results and insufficient data.

Quality assessment and data extraction

The Newcastle-Ottawa Scale checklist was used to assess the quality of the studies. The required data, including the first author, country, population sample size, number of H. pylori strains, diagnostic method, and frequency of cagA-positive strains, are listed in Table 1. All participants were divided into case (gastric MALT lymphoma) and control (gastritis or non-ulcer dyspepsia) groups.

Table 1

Characteristics of included studies

First author

Year

Country

Population size

H. pylori strains

Diagnostic method

cagA positive H. pylori strains

Gastritis or NUD

MALT

Jong et al.

1996

Netherlands

89

89

Culture-PCR

26

7

Peng et al.

1998

United kingdom

123

123

PCR

17

37

Lamarque et al.

1999

France

598

182

ELISA

20

10

Doorn et al.

1999

Netherlands

36

36

Culture-PCR

16

5

Schmaußer et al.

2000

Germany

30

30

ELISA

14

12

Delchier et al.

2001

France

598

162

ELISA

22

29

Koehler et al.

2003

Germany

121

91

Multiplex-PCR

27

15

Lehours et al.

2009

France

79

79

PCR

22

21

Talebi et al.

2013

Iran

134

128

Culture-PCR

63

5

Hashinaga et al.

2016

Japan

52

52

Culture-PCR

4

12

 

Statistical analysis

All statistical analyses were performed using Comprehensive Meta-Analysis software (Ver 2.2; Biostat, Englewood, NJ). The colonization rate of cagA-positive strains in both groups was reported as event rate (EER) with 95% confidence intervals (CIs). The impact of cagA gene status on the development of gastric MALT was also measured using the odds ratio (OR) at 95% CIs. The heterogeneity between studies was assessed with the I2 > 50 test and Cochran’s Q Statistic p value > 0.05. High levels of heterogeneity were evaluated according to the random-effects model with the DerSimonian and Laird method. In contrast, the fixed-effects model, based on the Mantel-Haenszel method, was used for low levels of heterogeneity. Furthermore, publication bias was assessed using asymmetry of funnel plots, Begg’s test p value, and Egger’s test p value.

Reults

Characteristics of selected studies

A total of 153 articles were collected in the initial search, and finally 10 eligible articles met our criteria and were included in the current analysis[3241]. A flowchart of the article search strategy and study selection is presented in Fig. 1.

All eligible studies were conducted from 1996–2016, and data from 1860 patients were reviewed in these studies. Two studies were performed on the Asian population, and eight studies were conducted in Western countries. No significant relationship was observed between cagA status and age/sex distribution in any of the studies. In two studies, the association between the cagA genotype and the development of gastric MALT lymphoma was controversial[33, 40]. Unfortunately, the association between CagA EPIYA motifs and gastric MALT lymphoma was not evaluated in all studies, so we could not investigate this association. However, in three studies, the association between cagA status and both low-grade and high-grade forms of gastric MALT lymphoma was investigated[33, 36, 37]. In addition, the association between vacA status and gastric MALT was assessed in six studies[3336, 39, 41].

Association between CagA status and susceptibility to gastric MALT

In this study, patients were divided into cases (gastric MALT lymphoma: 280 patients) and control (gastritis/non-ulcer dyspepsia (NUD): 414 patients). The prevalence of CagA-expressing strains in patients with MALT lymphoma and gastritis/NUD patients was estimated to be 54.6% (44–64.7 with 95% CIs) and 56.4% (41.5–70.3 with 95% CIs), respectively. However, according to the subgroup analysis by different geographical regions, we found that the frequency of cagA-positive strains in patients with gastritis/NUD in Western countries was higher than that in Asian countries (57.6% vs. 36%). The results showed that the cagA genotype was not significantly different between Western and Asian patients with gastric MALT lymphoma.

Based on the results of statistical analysis, we observed a weak correlation between cagA status and gastric MALT lymphoma (OR: 1.01; 0.71–1.42 with 95% CIs; I2: 83.52; Q-Value: 54.61; p value: 0.01; Egger’s p value: 0.36; Begg’s p value: 0.28). In the subgrouping, we found that there was an inverse significant association between cagA genotype and gastric MALT lymphoma in the Asian population (OR: 0.10; 0.03–0.31 with 95% CIs; I2: 95.6; Q-Value: 23.08; p value: 0.08), while there was a marginal association between cagA and the development of gastric MALT lymphoma in Western countries (OR: 1.30; 0.90–1.87 with 95% CIs; I2: 45.83; Q-Value: 12.92; p value: 0.58). Surprisingly, we observed a strong positive significant association between cagA status and the development of high-grade gastric MALT lymphoma (OR: 6.43; 2.45–16.84 with 95% CIs; I2: 0.00; Q-Value: 0.6; p value: 0.73; Egger’s p value: 0.21; Begg’s p value: 0.21).

Unfortunately, no studies have yet examined the relationship between CagA and VacA in the ‎development of MALT lymphoma, but VacA induces apoptosis by forming a vacuole ‎and release of cytochrome c from mitochondria and appears to inhibit the development of ‎MALT lymphoma[42, 43]. We found that there was a significant inverse association between infection with VacA-expressing H. pylori strains and gastric MALT lymphoma (OR: 0.92; 0.57–1.50 with 95% CI; I2: 32.3; Q-Value: 7.39; p value: 0.1; Egger’s p value: 0.79; Begg’s p value: 0.45). Therefore, despite the small sample size and significant heterogeneity between the studies, the results of the present study showed that cagA status has a marginal association with the risk of gastric MALT lymphoma, especially in Western countries. We also observed a strong association between CagA status and the development of high-grade MALT, indicating the importance of this virulence factor in the immune-pathogenesis of gastric MALT lymphoma. However, further investigation is required to confirm the results of this study.

Publication bias analysis

In the present study, the presence of bias in publication was evaluated using Begg’s p value and Egger’s p value. We did not observe any significant publication bias in the present study, although the funnel plot showed a slight publication bias in the eligible studies.

Discussion

MALT lymphoma refers to a group of low-grade B-cell lymphomas that involve the extranodal region of the mucosal and non-mucosal organs. Surprisingly, these organs normally lack lymphocytes, and infiltration of B-cell lymphocytes occurs as a result of chronic inflammation or autoimmune diseases (particularly Hashimoto’s thyroiditis). Gastric MALT lymphoma is the most common marginal zone lymphoma[44, 45]. Although almost 80% of low-grade MALT lymphomas are associated with H. pylori infection, disorders such as trisomy 3, trisomy 18, deletion in p16, and specific chromosomal translocations, such as t(1; 14)(p22; q32) and t(11; 18)(q21; q21) also increase the risk of gastric MALT lymphoma in H. pylori-negative individuals[46]. Today, the role of H. pylori infection in the development of gastric MALT lymphoma has been well-described, and recent studies have shown that eradication infection in low-grade lymphomas can lead to lymphoma regression in 60–90% of cases[47]. The most likely hypothesis is that persistent infection with H. pylori can lead to inflammation and infiltration of lymphocytes into the stomach with persistent stimulation of the immune response, and active proliferation of B cells leads to the formation of lymph follicles and the onset of gastric MALT lymphoma[48]. To date, several studies have attempted to investigate the association between cagA status and the development of gastric MALT lymphoma, but the results are unclear[3133]. In addition, no comprehensive meta-analysis was performed in this area; therefore, using the available evidence, we performed the present meta-analysis to evaluate the exact role of the CagA antigen in the development of gastric MALT lymphoma.

Our results showed that there is a marginal association between cagA status and gastric MALT lymphoma. In the subgroup analysis, we observed a weak association between CagA and MALT lymphoma in Western countries. Interestingly, the present analysis showed an inverse association between cagA status and gastric MALT lymphoma risk in the Asian population (OR: 0.10; 0.03–0.31 with 95% CIs).

Previous studies indicated that early lymphomagenesis in lymphomas is a process related to CD4 + T cells stimulated by H. pylori antigens, and the proliferation of B-cell gastric lymphoma is dependent on CD40-mediated signaling, Th2 activities, co-stimulatory CD80, and CD86[24, 4952]. Hussel et al. (1993) in their studies showed that the reduction of infiltrating T cells can significantly disrupt the effect of H. pylori infection on tumor B-cell proliferation[53]. Umehara et al. found that CagA could inhibit B-lymphoid cell proliferation by IL-3-dependent signaling by targeting the JAK-STAT pathway[26]. Furthermore, recent studies have shown an association between t(11; 18)(q21; q21) and infection with cagA-positive H. pylori strains in gastric MALT lymphoma; the anti-CagA titer is significantly higher in people with t(11; 18)(q21; q21)[46]. Liu et al. (2001) showed that the API2–MALT1 chimeric transcript was observed in all cases of H. pylori-infected gastric MALT lymphoma[54]. However, there is no correlation between H. pylori infection and the presence of API2–MALT1[55]. Thus, the formation of gastric MALT lymphoma appears to be more dependent on H. pylori antigen stimulation and T cell-mediated response. H. pylori cagA-positive strains with risk factors such as t(11; 18)(q21; q21) or API2–MALT1 chimeric translocation, and suppression of p53 accumulation as a cofactor, can effectively contribute to progression of stomach MALT lymphoma[26, 56].

Ohnishi et al. (2008) demonstrated the major role of CagA in the development of gastric and hematologic neoplasms[57]. After transfer to B-cell lymphocytes via the type 4 secretory system (T4SS), CagA, through the formation of phosphorylated CagA-SHP-2 complex by affecting ERK1, ERK2, p38MAPKs, ‎BCL2, and NF-κB, as well as accumulation of p53 or inhibition of the JAK-STAT signaling pathway, promotes lymphogenesis and immortalization of B-cell lymphocytes‎[26, 48, 58]. Gastric MALT lymphoma is classified into two subclasses based on the percentage of blast cells, including low-grade and high-grade lymphomas. Evaluation of cagA status in patients with low-grade and high-grade gastric lymphomas showed that this gene significantly increases the risk of developing high-grade lymphoma (OR: 6.43; 2.45–16.84 with 95% CIs). Based on previous studies, the presence of cagA-positive H. pylori strains in patients with high-grade lymphomas is significantly higher than that in patients with low-grade lymphomas[31]. The role of VacA in gastric MALT lymphoma is also controversial, and in one study, Miehlke et al. (1998) showed that the level of the vacA s1m1 genotype in gastric MALT lymphoma patients is high; however, ‎Doorn et al. (1999) rejected this hypothesis‎[35, 59]. Although we could not assess the correlation between cagA and vacA, we observed an inverse association between the vacA genotype and gastric MALT lymphoma (OR: 0.92; 0.57–1.50 with 95% CIs). Although vacA is a potent immune gene, given the fact that this protein causes apoptosis, it does not appear to play a significant role in the development of gastric MALT lymphoma[60, 61]. In general, the most likely hypothesis to describe the role of H. pylori in the development of ‎gastric MALT lymphoma is that this bacterium (CagA-dependent or independent) causes ‎chronic gastritis, resulting in the production of IL-8 and other molecules associated with neutrophil chemotaxis. Neutrophil activation leads to destruction of the gastric mucosa and close contact of CD4 + T cells with H. pylori, where the activity of DC and CD4 + T cells causes B cells to mature. Continuous stimulation and proliferation of B-cell lymphocytes leads to the formation of lymph follicles, in which case the patient develops low-grade lymphomas. In addition, no H. pylori eradication, particularly of cagA-positive strains, leads to the translocation of CagA into B cells. Intracellular CagA causes DNA and microRNA damage by reactive oxygen and nitrogen species, inhibition of p53, and chromosomal translocation, and ultimately the development of high-grade lymphomas (Fig. 2).

Our study had several limitations including: I) low sample size, II) evaluation of only studies published in English, III) high heterogeneity in some cases, IV) inaccessibility to the raw data for finding EPIYA motifs, correlation of CagA and VacA. The results of the study are unstable under the influence of significant heterogeneity, and further research is needed for confirmation.

Conclusion

In the present study, we performed a large pooled analysis to evaluate the role of CagA in gastric MALT lymphoma. Based on the available findings, the role of CagA was marginal in the initiation of low-grade lymphomas, especially in Asian countries, where we observed an inverse association between CagA and low-grade gastric MALT lymphoma. However, CagA can stimulate lymphogenesis and lead to the contentious proliferation and immortalization of B cells; therefore, it plays an important role in the development of high-grade gastric MALT lymphoma.

Abbreviations

H. pylori: Helicobacter pylori

MALT: mucosa-associated lymphoid tissue lymphoma

CagA: cytotoxin-associated gene A

PCR: Polymerase Chain Reaction

Declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Availability of data and materials

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Funding

Not applicable.

Authors' Contributions

MK1 and AS equally contributed to the conception and design of the study; MK1 and MK2 performed the literature review and analysis. All authors equally contributed to drafting, critical revision, editing, and final approval.

Acknowledgements

Not applicable.

References

  1. Kusters JG, Van Vliet AH, Kuipers EJ. Pathogenesis of Helicobacter pylori infection. Clinical microbiology reviews 2006; 19(3): 449-490
  2. De Falco M, Lucariello A, Iaquinto S, Esposito V, Guerra G, De Luca A. Molecular mechanisms of Helicobacter pylori pathogenesis. Journal of cellular physiology 2015; 230(8): 1702-1707
  3. Youssefi M, Tafaghodi M, Farsiani H, Ghazvini K, Keikha M. Helicobacter pylori infection and autoimmune diseases; Is there an association with systemic lupus erythematosus, rheumatoid arthritis, autoimmune atrophy gastritis and autoimmune pancreatitis? A systematic review and meta-analysis study. Journal of Microbiology, Immunology and Infection 2020
  4. Hooi JK, Lai WY, Ng WK, Suen MM, Underwood FE, Tanyingoh D, Malfertheiner P, Graham DY, Wong VW, Wu JC. Global prevalence of Helicobacter pylori infection: systematic review and meta-analysis. Gastroenterology 2017; 153(2): 420-429
  5. Karbalaei M, Keikha M. Potential association between the hopQ alleles of Helicobacter pylori and gastrointestinal diseases: A systematic review and meta-analysis. Meta Gene 2020: 100816
  6. Šterbenc A, Jarc E, Poljak M, Homan M. Helicobacter pylori virulence genes. World journal of gastroenterology 2019; 25(33): 4870
  7. Servetas SL, Kim A, Su H, Cha JH, Merrell DS. Comparative analysis of the Hom family of outer membrane proteins in isolates from two geographically distinct regions: the United States and South Korea. Helicobacter 2018; 23(2): e12461
  8. Gressmann H, Linz B, Ghai R, Pleissner K-P, Schlapbach R, Yamaoka Y, Kraft C, Suerbaum S, Meyer TF, Achtman M. Gain and loss of multiple genes during the evolution of Helicobacter pylori. PLoS Genet 2005; 1(4): e43
  9. Higashi H, Tsutsumi R, Fujita A, Yamazaki S, Asaka M, Azuma T, Hatakeyama M. Biological activity of the Helicobacter pylori virulence factor CagA is determined by variation in the tyrosine phosphorylation sites. Proceedings of the National Academy of Sciences 2002; 99(22): 14428-14433
  10. Lind J, Backert S, Hoffmann R, Eichler J, Yamaoka Y, Perez-Perez GI, Torres J, Sticht H, Tegtmeyer N. Systematic analysis of phosphotyrosine antibodies recognizing single phosphorylated EPIYA-motifs in CagA of East Asian-type Helicobacter pylori strains. BMC microbiology 2016; 16(1): 201
  11. Li Q, Liu J, Gong Y, Yuan Y. Association of CagA EPIYA-D or EPIYA-C phosphorylation sites with peptic ulcer and gastric cancer risks: A meta-analysis. Medicine 2017; 96(17)
  12. Yamaoka Y. Mechanisms of disease: Helicobacter pylori virulence factors. Nature reviews Gastroenterology & hepatology 2010; 7(11): 629
  13. Matsunari O, Shiota S, Suzuki R, Watada M, Kinjo N, Murakami K, Fujioka T, Kinjo F, Yamaoka Y. Association between Helicobacter pylori virulence factors and gastroduodenal diseases in Okinawa, Japan. Journal of clinical microbiology 2012; 50(3): 876-883
  14. Crabtree J, Xiang Z, Lindley I, Tompkins D, Rappuoli R, Covacci A. Induction of interleukin-8 secretion from gastric epithelial cells by a cagA negative isogenic mutant of Helicobacter pylori. Journal of clinical pathology 1995; 48(10): 967-969
  15. Crabtree J, Covacci A, Farmery S, Xiang Z, Tompkins D, Perry S, Lindley I, Rappuoli R. Helicobacter pylori induced interleukin-8 expression in gastric epithelial cells is associated with CagA positive phenotype. Journal of clinical pathology 1995; 48(1): 41-45
  16. Machado AMD, Figueiredo C, Touati E, Maximo V, Sousa S, Michel V, Carneiro F, Nielsen FC, Seruca R, Rasmussen LJ. Helicobacter pylori infection induces genetic instability of nuclear and mitochondrial DNA in gastric cells. Clinical Cancer Research 2009; 15(9): 2995-3002
  17. Isaacson P, Wright DH. Malignant lymphoma of mucosa‐associated lymphoid tissue. A distinctive type of B‐cell lymphoma. Cancer 1983; 52(8): 1410-1416
  18. Nakamura S, Matsumoto T. Helicobacter pylori and gastric mucosa-associated lymphoid tissue lymphoma: recent progress in pathogenesis and management. World Journal of Gastroenterology: WJG 2013; 19(45): 8181
  19. Carlson SJ, Yokoo H, Vanagunas A. Progression of gastritis to monoclonal B-cell lymphoma with resolution and recurrence following eradication of Helicobacter pylori. Jama 1996; 275(12): 937-939
  20. Bilgilier C, Simonitsch-Klupp I, Kiesewetter B, Raderer M, Dolak W, Makristathis A, Steininger C. Prevalence of clarithromycin-resistant Helicobacter pylori strains in gastric mucosa-associated lymphoid tissue lymphoma patients. Annals of hematology 2016; 95(7): 1115-1120
  21. Asenjo L, Gisbert J. Prevalence of Helicobacter pylori infection in gastric MALT lymphoma: a sistematic review. Revista Española de Enfermedades Digestivas 2007; 99(7): 398
  22. Wotherspoon AC, Diss T, Pan L, Isaacson P, Doglioni C, Moschini A, de Boni M. Regression of primary low-grade B-cell gastric lymphoma of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori. The Lancet 1993; 342(8871): 575-577
  23. Greiner A, Knörr C, Qin Y, Sebald W, Schimpl A, Banchereau J, Müller-Hermelink H. Low-grade B cell lymphomas of mucosa-associated lymphoid tissue (MALT-type) require CD40-mediated signaling and Th2-type cytokines for in vitro growth and differentiation. The American journal of pathology 1997; 150(5): 1583
  24. HUSSELL T, ISAACSON PG, CRABTREE JE, Spencer J. Helicobacter pylori‐specific tumour‐infiltrating T cells provide contact dependent help for the growth of malignant B cells in low‐grade gastric lymphoma of mucosa‐associated lymphoid tissue. The Journal of pathology 1996; 178(2): 122-127
  25. Lin W-C, Tsai H-F, Kuo S-H, Wu M-S, Lin C-W, Hsu P-I, Cheng A-L, Hsu P-N. Translocation of Helicobacter pylori CagA into Human B lymphocytes, the origin of mucosa-associated lymphoid tissue lymphoma. Cancer research 2010; 70(14): 5740-5748
  26. Umehara S, Higashi H, Ohnishi N, Asaka M, Hatakeyama M. Effects of Helicobacter pylori CagA protein on the growth and survival of B lymphocytes, the origin of MALT lymphoma. Oncogene 2003; 22(51): 8337-8342
  27. Chen CY, Wang FY, Wan HJ, Jin XX, Wei J, Wang ZK, Liu C, Lu H, Shi H, Li DH. Amino acid polymorphisms flanking the EPIYA‐A motif of H elicobacter pylori CagA C‐terminal region is associated with gastric cancer in E ast C hina: Experience from a single center. Journal of digestive diseases 2013; 14(7): 358-365
  28. Krisch LM, Posselt G, Hammerl P, Wessler S. CagA phosphorylation in Helicobacter pylori-infected B cells is mediated by the nonreceptor tyrosine kinases of the Src and Abl families. Infection and immunity 2016; 84(9): 2671-2680
  29. Peng JC, Zhong L, Ran ZH. Primary lymphomas in the gastrointestinal tract. Journal of digestive diseases 2015; 16(4): 169-176
  30. Wang H-P, Zhu Y-L, Shao W. Role of Helicobacter pylori virulence factor cytotoxin-associated gene A in gastric mucosa-associated lymphoid tissue lymphoma. World journal of gastroenterology: WJG 2013; 19(45): 8219
  31. Eck M, Schmausser B, Haas R, Greiner A, Czub S, Muller-Hermelink H. MALT-type lymphoma of the stomach is associated with Helicobacter pylori strains expressing the CagA protein. Gastroenterology 1997; 112(5): 1482-1486
  32. De Jong D, Van Der Hulst RW, Pals G, Van Dijk WC, Van Der Ende A, Tytgat GN, Taal BG, Boot H. Gastric non-Hodgkin lymphomas of mucosa-associated lymphoid tissue are not associated with more aggressive Helicobacter pylori strains as identified by CagA. American journal of clinical pathology 1996; 106(5): 670-675
  33. Peng H, Ranaldi R, Diss TC, Isaacson PG, Bearzi I, Pan L. High frequency of CagA+ Helicobacter pylori infection in high‐grade gastric MALT B‐cell lymphomas. The Journal of Pathology: A Journal of the Pathological Society of Great Britain and Ireland 1998; 185(4): 409-412
  34. Lamarque D, Gilbert T, Roudot-Thoraval F, Deforges L, Chaumette MT, Delchier JC. Seroprevalence of eight Helicobacter pylori antigens among 182 patients with peptic ulcer, MALT gastric lymphoma or non-ulcer dyspepsia. Higher rate of seroreactivity against CagA and 35-kDa antigens in patients with peptic ulcer originating from Europe and Africa. European journal of gastroenterology & hepatology 1999; 11(7): 721-726
  35. van Doorn NE, Namavar F, van Doorn L-J, Durrani Z, Kuipers EJ, Vandenbroucke-Grauls CM. Analysis of vacA, cagA, and IS605 Genotypes and Those Determined by PCR Amplification of DNA between Repetitive Sequences of Helicobacter pyloriStrains Isolated from Patients with Nonulcer Dyspepsia or Mucosa-Associated Lymphoid Tissue Lymphoma. Journal of clinical microbiology 1999; 37(7): 2348-2349
  36. Schmaußer B, Eck M, Greiner A, Kraus M, Müller-Hermelink HK. Mucosal humoral immune response to CagA shows a high prevalence in patients with gastric MALT-type lymphoma. Virchows Archiv 2000; 436(2): 115-118
  37. Delchier J-C, Lamarque D, Levy M, Tkoub EM, Copie-Bergman C, Deforges L, Chaumette M-T, Haioun C. Helicobacter pylori and gastric lymphoma: high seroprevalence of CagA in diffuse large B-cell lymphoma but not in low-grade lymphoma of mucosa-associated lymphoid tissue type. The American journal of gastroenterology 2001; 96(8): 2324-2328
  38. Koehler C, Mues M, Dienes H, Kriegsmann J, Schirmacher P, Odenthal M. Helicobacter pylori genotyping in gastric adenocarcinoma and MALT lymphoma by multiplex PCR analyses of paraffin wax embedded tissues. Molecular Pathology 2003; 56(1): 36
  39. Lehours P, Zheng Z, Skoglund A, Mégraud F, Engstrand L. Is there a link between the lipopolysaccharide of Helicobacter pylori gastric MALT lymphoma associated strains and lymphoma pathogenesis? PLoS One 2009; 4(10): e7297
  40. Abadi ATB, Ghasemzadeh A, Mobarez AM. Low frequency of cagA-positive Helicobacter pylori strains isolated from Iranian patients with MALT lymphoma. Internal and emergency medicine 2013; 8(1): 49-53
  41. Hashinaga M, Suzuki R, Akada J, Matsumoto T, Kido Y, Okimoto T, Kodama M, Murakami K, Yamaoka Y. Differences in amino acid frequency in CagA and VacA sequences of Helicobacter pylori distinguish gastric cancer from gastric MALT lymphoma. Gut pathogens 2016; 8(1): 54
  42. Matsumoto A, Isomoto H, Nakayama M, Hisatsune J, Nishi Y, Nakashima Y, Matsushima K, Kurazono H, Nakao K, Hirayama T. Helicobacter pylori VacA reduces the cellular expression of STAT3 and pro-survival Bcl-2 family proteins, Bcl-2 and Bcl-X L, leading to apoptosis in gastric epithelial cells. Digestive diseases and sciences 2011; 56(4): 999-1006
  43. Palframan SL, Kwok T, Gabriel K. Vacuolating cytotoxin A (VacA), a key toxin for Helicobacter pylori pathogenesis. Frontiers in cellular and infection microbiology 2012; 2: 92
  44. Filip PV, Cuciureanu D, Diaconu LS, Vladareanu AM, Pop CS. MALT lymphoma: epidemiology, clinical diagnosis and treatment. Journal of Medicine and Life 2018; 11(3): 187
  45. Ye H, Liu H, Attygalle A, Wotherspoon AC, Nicholson AG, Charlotte Fdr, Leblond V, Speight P, Goodlad J, Lavergne-Slove A. Variable frequencies of t (11; 18)(q21; q21) in MALT lymphomas of different sites: significant association with CagA strains of H pylori in gastric MALT lymphoma. Blood 2003; 102(3): 1012-1018
  46. Sumida T, Kitadai Y, Hiyama T, Shinagawa K, Tanaka M, Kodama M, Masuda H, Ito M, Tanaka S, Yoshihara M. Antibodies to Helicobacter pylori and CagA protein are associated with the response to antibacterial therapy in patients with H. pylori‐positive API2–MALT1‐negative gastric MALT lymphoma. Cancer science 2009; 100(6): 1075-1081
  47. Zucca E, Roggero E, Cavalli F. Helicobacter pylori eradication in gastric: mucosa-associated lymphoid: tissue lymphomas. Annals of internal medicine 1996; 124(2): 275-276
  48. Floch P, Mégraud F, Lehours P. Helicobacter pylori strains and gastric MALT lymphoma. Toxins 2017; 9(4): 132
  49. Farinha P, Gascoyne RD. Helicobacter pylori and MALT lymphoma. Gastroenterology 2005; 128(6): 1579-1605
  50. Hauer A, Finn T, MacDonald T, Spencer J, Isaacson P. Analysis of TH1 and TH2 cytokine production in low grade B cell gastric MALT-type lymphomas stimulated in vitro with Helicobacter pylori. Journal of clinical pathology 1997; 50(11): 957-959
  51. Knörr C, Amrehn C, Seeberger H, Rosenwald A, Stilgenbauer S, Ott G, Hermelink H-KM, Greiner A. Expression of costimulatory molecules in low-grade mucosa-associated lymphoid tissue-type lymphomas in vivo. The American journal of pathology 1999; 155(6): 2019-2027
  52. Mueller A, O'Rourke J, Chu P, Chu A, Dixon MF, Bouley DM, Lee A, Falkow S. The role of antigenic drive and tumor-infiltrating accessory cells in the pathogenesis of helicobacter-induced mucosa-associated lymphoid tissue lymphoma. The American journal of pathology 2005; 167(3): 797-812
  53. Hussell T, Isaacson PG, Spencer J, Crabtree J. The response of cells from low-grade B-cell gastric lymphomas of mucosa-associated lymphoid tissue to Helicobacter pylori. The Lancet 1993; 342(8871): 571-574
  54. Liu H, Ruskon-Fourmestraux A, Lavergne-Slove A, Ye H, Molina T, Bouhnik Y, Hamoudi RA, Diss TC, Dogan A, Megraud F. Resistance of t (11; 18) positive gastric mucosa-associated lymphoid tissue lymphoma to Helicobacter pylori eradication therapy. The Lancet 2001; 357(9249): 39-40
  55. Baens M, Maes B, Steyls A, Geboes K, Marynen P, De Wolf-Peeters C. The product of the t (11; 18), an API2-MLT fusion, marks nearly half of gastric MALT type lymphomas without large cell proliferation. The American journal of pathology 2000; 156(4): 1433-1439
  56. Kuo S, Chen L, Lin C, Wu M, Hsu P, Tsai H, Chu C, Tzeng Y, Wang H, Yeh K. Detection of the Helicobacter pylori CagA protein in gastric mucosa-associated lymphoid tissue lymphoma cells: clinical and biological significance. Blood cancer journal 2013; 3(7): e125
  57. Ohnishi N, Yuasa H, Tanaka S, Sawa H, Miura M, Matsui A, Higashi H, Musashi M, Iwabuchi K, Suzuki M. Transgenic expression of Helicobacter pylori CagA induces gastrointestinal and hematopoietic neoplasms in mouse. Proceedings of the National Academy of Sciences 2008; 105(3): 1003-1008
  58. Zhu Y, Wang C, Huang J, Ge Z, Dong Q, Zhong X, Su Y, Zheng S. The Helicobacter pylori virulence factor CagA promotes Erk1/2‐mediated Bad phosphorylation in lymphocytes: a mechanism of CagA‐inhibited lymphocyte apoptosis. Cellular microbiology 2007; 9(4): 952-961
  59. Miehlke S, Meining A, Morgner A, Bayerdörffer E, Lehn N, Stolte M, Graham DY, Go MF. Frequency of vacA genotypes and cytotoxin activity in Helicobacter pylori associated with low-grade gastric mucosa-associated lymphoid tissue lymphoma. Journal of clinical microbiology 1998; 36(8): 2369-2370
  60. Ihan A, Pinchuk IV, Beswick EJ. Inflammation, Immunity, and Vaccines for H elicobacter pylori Infection. Helicobacter 2012; 17: 16-21
  61. Cover TL, Krishna US, Israel DA, Peek RM. Induction of gastric epithelial cell apoptosis by Helicobacter pylori vacuolating cytotoxin. Cancer research 2003; 63(5): 951-957.