Skip to main content
  • Research article
  • Open access
  • Published:

Prognostic significance of helicobacter pylori-infection in gastric diffuse large B-cell lymphoma

Abstract

Background

Helicobacter pylori (H. pylori) is thought to have an oncogenic effect on the development of gastric malignancies. However, the effect of H. pylori status on the prognosis of gastric diffuse large B-cell lymphoma (DLBCL) remains unconfirmed. This study aimed to identify the prognostic importance of H. pylori infection in de novo gastric DLBCL.

Methods

One hundred and twenty-nine patients diagnosed with primary de novo gastric DLBCL at the West China Hospital of Sichuan University from 1st January 2009 to 31st May 2016 were included. The clinical features of the patients were documented. H. pylori status was assessed via urease breath tests and histologic examinations. The prognostic value of H. pylori was verified via univariate and multivariate analyses.

Results

Over a median follow-up of 52.2 months (range 4–116), the 5-year overall survival (OS) for all patients was 78.7%. Patients with H. pylori infections had significantly better 5-year PFS and OS than did the H. pylori-negative subgroup (5-year PFS, 89.3% vs. 74.1%, P = 0.040; 5-year OS, 89.7% vs. 71.8%, P = 0.033). Negative H. pylori status and poor ECOG performance were independent negative prognostic indicators for both PFS and OS (PFS, P = 0.045 and P = 0.001, respectively; OS, P = 0.021 and P < 0.001, respectively).

Conclusions

H. pylori status in de novo gastric DLBCL can be a promising predictor of disease outcome, and patients with negative H. pylori status require careful follow-up since they tend to have a worse outlook.

Peer Review reports

Background

The gastrointestinal tract is a common site of extranodal non-Hodgkin lymphoma (NHL), with the stomach being the affected site in 60% of all NHL patients with digestive tract involvement [1, 2]. Among gastric lymphomas, mucosa-associated lymphoid tissue (MALT) lymphoma and diffuse large B-cell lymphoma (DLBCL) are the two most common types [3]. DLBCL in the stomach is a heterogeneous disease, and it is usually divided into two categories: DLBCL with features of MALT lymphoma (DLBCL (MALT)) and DLBCL without evidence of MALT (de novo DLBCL or pure DLBCL) [4]. Up to 20% of patients with DLBCL have been identified as a concurrent component of MALT [5].

Helicobacter pylori (H. pylori), a spiral-shaped, micro-aerophilic bacterium that inhabits the human stomach, is estimated to colonize more than half of the world’s human population [6]. The bacterium has been categorized as a class I carcinogen, giving rise to a new method for classifying gastric carcinoma [7]. Previous studies showed that H. pylori infections can induce a gastric lymphoid tissue response and that it might be an oncogenic factor during the development of malignant gastric lymphomas, including MALT lymphoma and DLBCL [8,9,10]. However, accumulating evidence has proven that H. pylori-positive status can support long-term survival and lead to better prognoses in gastric carcinoma patients [11,12,13]. It is thought that DLBCL (MALT) is independent of H. pylori status, as it fails to respond to antibiotic therapy according to the WHO (World Health Organization) classification and contains a component of high-grade transformed MALT lymphoma that differs from low-grade and H. pylori-dependent MALT lymphomas (MALT lymphoma) [5, 14,15,16]. However, numerous studies have found that a substantial percentage of gastric DLBCL (MALT) is associated with H. pylori infection and that it responds effectively to H. pylori eradication, especially early-stage DLBCL (MALT) [17, 18]. Of note, de novo DLBCL, the pathogenesis of which was once thought to be different from that of MALT lymphoma, has also been proven to correlate with H. pylori status based on limited data [19, 20]. Until now, this clinically relevant finding has not been validated in large studies at other medical centers.

In this retrospective study, we evaluated the prognostic value of H. pylori status for patients with de novo DLBCL, especially among the early-stage population, at a larger medical center.

Methods

Patients: diagnosis and treatment

A retrospective review of all patients with the diagnosis of primary de novo DLBCL of the stomach at West China Hospital of Sichuan University from 1st January 2009 to 31st May 2016 was performed. Pathological specimens were obtained from both endoscopic biopsies and surgical resections, and diagnosis was based on the World Health Organization (WHO) classification system for hematologic malignancies [16]. Tumors without histological features of MALT lymphoma, including dense infiltration of centrocyte-like cells in the lamina propria and typical lymphoepithelial lesions [21, 22], were classified as de novo DLBCL. Only patients with primary involvement of stomach or with predominant gastric lesions were included. Patients with secondary gastric lymphoma or evidence of MALT origin were excluded. Paraffin-embedded, formalin-fixed tumor specimens where immunohistochemically stained for CD20, CD3, CD5, CD10, BCL6 and MUM1 (Fig. 1).

Fig. 1
figure 1

An example of immunohistochemical features of germinal center B-cell de novo gastric DLBCL. a diffuse large cells infiltrating the mucosa (hematoxylin-eosin (H&E) staining, × 400), (b) positive CD20 expression, (c) positive BCL6 expression, (d) negative expression of CD10, (e) negative expression of MUM1, and (f) Warthin-Starry staining of H. pylori

Patients were treated with the following therapeutic modalities singly or in combination: surgery, chemotherapy or radiotherapy. Chemotherapy referred to CHOP (cyclophosphamide, doxorubicin, vincristine and prednisone) or CHOP-like regimens, with or without rituximab (R).

H. pylori infection

The status of H. pylori infection was confirmed at the baseline for each case via the results of at least one of two tests: histologic examination or a 14C-urea breath test (14C -UBT). The histologic examination consisted of Warthin-Sharry (W-S) staining of tissue specimens. The results of the 14C-UBT were reported as 14C disintegrations per minute (dpm), and dpm ≥ 100 and dpm < 100 were interpreted as positive and negative, respectively. Of note, all H. pylori-positive patients received antibiotic therapy against H. pylori, including bismuth compounds, proton pump inhibitor (PPI) and two of the following antibiotics: clarithromycin, amoxicillin, metronidazole or tetracycline.

Clinical evaluation

Staging workups included the collection of a detailed medical history, a physical examination that included examination of the peripheral lymph nodes and Waldeyer’s ring, complete hematological biochemical examinations, including serum lactate dehydrogenase (LDH), computed tomography (CT), gastric endoscopy, endoscopic ultrasonography, positron emission tomography (PET)/CT, and bone marrow aspiration and biopsy. For patients who received surgical interventions, the intraoperative exploration and pathological results were further considered. The staging and classification of the lesions were based on the Lugano staging system [23]. Furthermore, the performance status of each patient was assessed according to the Eastern Cooperative Oncology Group (ECOG) scale and the International Prognostic Index (IPI).

Statistical analyses

Pearson’s chi-squared test and Fisher’s exact test were used to evaluate the relationships between H. pylori status and other prognostic indicators and clinicopathological characteristics. Analyses were performed using follow-up data available on 31st May 2018. The primary endpoints of the current study were progression-free survival (PFS) and overall survival (OS). PFS was defined as the date of initial diagnosis until disease progression, relapse or death due to any cause. OS was measured from the date of diagnosis to the date of death from any cause or the date of a survivor’s final follow-up. Survival curves were estimated via the Kaplan-Meier method, and the differences between survival curves were compared via the log-rank test. All of the significant variables in the univariate analyses were included in multivariate analyses to evaluate the independent risk factors for PFS and OS via the Cox proportional hazards regression model. SPSS version 21.0 was used for the data analyses. P-values less than 0.05 were considered statistically significant. Our team has reviewed the data and reached an agreement on this final version.

Results

Baseline characteristics

The demographic baseline data and clinicopathologic parameters of the 129 included patients with de novo gastric DLBCL are listed in Table 1. The median age was 55 years old with a range of 21–84. 66 (51.2%) of the patients were women. Among the 129 patients, 64 (49.6%) were negative for H. pylori and 65 (50.4%) were positive for H. pylori. 71 (86.6%) patients had lesions in the antrum or corpus of the stomach, and B symptoms appeared in 45 (34.9%) patients. Most patients (> 50%) were early-stage and had better performance based on Lugano stage, IPI score and ECOG scale. Of note, the distribution of Lugano stage, IPI risk group, resection surgical treatment and LDH level were significantly associated with the H. pylori-positive and -negative subgroups (P < 0.05). Patients with H. pylori infections tended to remain in an early-stage of de novo DLBCL, whereas patients in the H. pylori-negative subgroup had relatively poor performance. The remaining clinical variables were similar in both H. pylori subgroups (P > 0.05).

Table 1 Association between clinicopathologic features and H. pylori status of 129 patients with de novo gastric diffuse large B-cell lymphoma

Over a median follow-up of 52.2 months (range 4–116), the 5-year OS for all patients was 78.7%. The overall median PFS was 91.8 months (95% confidence interval [CI] 83.8–99.7), and the median overall survival (OS) was 96.0 months (95% CI 88.9–103.0). Patients with H. pylori-positive de novo gastric DLBCL had significantly better 5-year PFS and OS than patients in the H. pylori-negative subgroup (5-year PFS, 89.3% vs. 74.1%, P = 0.040; 5-year OS, 89.7% vs. 71.8%, P = 0.033) (Tables 2 and 3).

Table 2 Univariate and multivariate analysis of prognostic factors for progression-free survival in 129 patients with de novo gastric diffuse large B-cell lymphoma
Table 3 Univariate and multivariate analysis of prognostic factors for overall survival in 129 patients with de novo gastric diffuse large B-cell lymphoma

Univariate and multivariate analyses

We next investigated the associations between important clinicopathologic parameters and patient survival via a Cox proportional hazard regression analysis (Tables 2 and 3).

Upon follow-up, 28 patients showed disease progression (22 were H. pylori-negative and 6 were H. pylori-positive). The mean PFS was 100.6 months (95% CI 92.0–109.2) in patients positive for H. pylori, compared with 85.0 months (95% CI 74.2–95.7) for patients in the H. pylori-negative subgroup (P = 0.040). The presence of B symptoms (P = 0.012), advanced Lugano stage (P = 0.013), poor ECOG performance status (P < 0.001), classification into higher IPI risk groups (P < 0.001), lack of resection surgery (P = 0.044) and negative H. pylori status (P = 0.040) were significantly associated with poor PFS of de novo gastric DLBCL in the univariate analyses (Table 2). The multivariate analyses revealed that the presence of B symptoms (hazard ratio = 2.605; P = 0.004) and poor ECOG performance status (hazard ratio = 5.002; P = 0.001) could independently predict poor PFS outcomes of de novo gastric DLBCL. However, positive H. pylori status was a positive predictor of PFS (hazard ratio = 0.379; P = 0.045) (Table 2).

Among the 25 patients who died during the follow-up, 5 were H. pylori-positive and the remained were H. pylori-negative. The mean OS was 102.7 months (95% CI 94.9–110.5) in H. pylori-positive patients, compared with 89.3 months (95% CI 79.4–99.3) in the patients in the H. pylori-negative subgroup (P = 0.033). Univariate analyses showed significant associations between OS and age (P = 0.033), Lugano staging (P = 0.012), ECOG performance (P < 0.001), IPI risk group (P < 0.001), resection surgery (P = 0.021), and H. pylori infection (P = 0.020) (Table 3). Multivariate analyses revealed that ECOG performance (hazard ratio = 6.216; P < 0.001) was an independent prognostic index for poor OS in de novo gastric DLBCL. Resection surgery (hazard ratio = 0.273; P = 0.003) and H. pylori infection (hazard ratio = 0.292; P = 0.021) were positive prognostic factors for OS (Table 3).

Kaplan-Meier survival analysis

Kaplan-Meier survival curves showed that H. pylori infection status (P = 0.025), resection surgery (P = 0.014), age (P = 0.027), ECOG performance status (P < 0.001), IPI risk group (P < 0.001) and Lugano stage (P = 0.008) were significantly associated with OS of de novo DLBCL (Fig. 2).

Fig. 2
figure 2

Effects of H. pylori status, surgery, age, ECOG performance, IPI risk group, and Lugano stage on overall survival of patients with de novo gastric diffuse large B-cell lymphoma according to Kaplan-Meier analysis. a Positive H. pylori status (p = 0.025), (b) Surgery (p = 0.014), (c) Age < 60 (p = 0.027), (d) ECOG< 2 (p < 0.001), (e) low and low intermediate risk (p < 0.001) and (f) Lugano stage I and II (p = 0.008) are positive prognostic factors for de novo gastric DLBCL

Subgroup analysis

Of note, in stage-specific analyses, H. pylori infection presented a significant association with OS in stage I or II (P = 0.034) but not in stage IIE or IV (P = 0.675) (Fig. 3).

Fig. 3
figure 3

Effects of H. pylori status according to Lugano stage on overall survival of patients with de novo gastric diffuse large B-cell lymphoma. Positive H. pylori status is associated with better prognosis in patients of (a) Lugano stage I and II rather than those of (b) stage IIE and IV (p = 0.034 and p = 0.675, respectively)

Discussion

Gastric MALT lymphoma has been claimed to be strongly associated with H. pylori infection, and eradication of H. pylori (HPE) even without oncological treatment has been used as the first-line treatment for this disease, especially for early stage MALT lymphoma [24, 25]. Gastric DLBCL (MALT) results from high-grade transformation of MALT lymphoma and is independent of H. pylori infection; however, previous studies have also proven that H. pylori eradication can lead to complete remission in a certain fraction of patients with H. pylori-positive DLBCL (MALT) [26]. Researchers have also found that some de novo DLBCL patients also presented complete remission after H. pylori eradication [27, 28]. Furthermore, an association between H. pylori infection and de novo DLBCL was validated in a large cohort [29]. Our results suggested that H. pylori-positive status was strongly indicative of better gastric de novo DLBCL prognosis.

H. pylori-induced lymphomagenesis is a multistep process involving H. pylori virulence factors (e.g., CagA, VacA and OipA), host factors and environmental conditions. Cytotoxin-associated gene A (CagA) protein, which is exposed on the surface of H. pylori cells via a type IV secretion system, is the most extensively studied H. pylori virulence factor. CagA can directly pass through the host membrane via an interaction with phosphatidylserine, after which it perturbs cell signaling in a way that can lead to oncogenesis [30]. Consistent with previous studies, our results indicated that despite the carcinogenic effect of H. pylori, H. pylori infection was associated with a less aggressive subtype of de novo DLBCL, and these patients showed better prognoses [19, 20]. Further subgroup analyses suggested that H. pylori infection was significantly associated with better survival outcomes in patients with early-stage gastric de novo DLBCL. As the identification of H. pylori status is possible during early cancer stages, it is useful for stratifying patients into risk groups and for predicting adverse disease outcomes; thus, its identification of as an independent predictor could establish it as a promising tool in clinical practice for helping to make treatment decisions.

One possible explanation for the critical role of H. pylori infection in improving the outlook of gastric de novo DLBCL is the immune cross-reactivity between H. pylori and the malignant B-cells of gastric DLBCL. Furthermore, previous studies have hypothesized that antigenic mimicry between H. pylori and the gastric mucosa might result in immune cross-reactions that affect tumor cells and suppress tumor progression as well as metastasis in gastric carcinoma [31, 32]. Furthermore, the immune response induced by H. pylori might also be cross-reactive against the malignant B-cells of gastric DLBCL since H. pylori can transfer CagA into both epithelial cells and B-lymphocytes and promote CagA expression in malignant B-cells [33]. Immune cross-reactions with malignant cells are characterized by the presence of mimic or absorbed H. pylori antigens, which results in improved survival outcomes in H. pylori-positive gastric DLBCL.

These newly uncovered underlying mechanisms demonstrate that H. pylori-positive de novo gastric DLBCL has less aggressive behavior. By examining genome-wide expression profiles of both mRNAs and miRNAs in H. pylori-positive and -negative de novo gastric DLBCL tissue specimens, a previous study confirmed that H. pylori infection is associated with a higher level of miR-200, which can inhibit Zinc-finger E-box-binding homeobox 1 (ZEB1) [20]. ZEB1 is expressed at a significantly higher level in DLBCL than that in reactive lymphoid tissue and is linked to an adverse prognosis in DLBCL [34]. ZEB1 promotes DLBCL progression via downregulation of BCL6, which is a known positive predictor for DLBCL [20]. However, the specific role of BCL6 in DLBCL progression requires further investigation. In light of its role in carcinogenesis, H. pylori might also affect other biological behaviors of gastric DLBCL, which may contribute to the survival benefits observed in the H. pylori-positive subgroup. Inherent differences in the tumors that develop secondary to H. pylori-infection and the carcinogenic functions of H. pylori infection require additional study.

This study is limited by its retrospective nature and single-institution bias. Furthermore, not all patients included in this study were treated with rigorous standard therapy. Some of the patients received chemotherapy without rituximab due to economic pressure, while other patients underwent surgery because they were either diagnosed with gastric DLBCL after surgery, or they were diagnosed years ago when surgery was still commonly performed. Despite the aforementioned limitations, to our knowledge, this study is the largest report examining the prognostic role of H. pylori in de novo gastric DLBCL.

Conclusions

In summary, patients with primary gastric de novo DLBCL without H. pylori infection are more likely to have poor prognoses than patients with the infection; therefore, the patients without H. pylori may benefit from more aggressive treatment and more systematic follow-up.

Availability of data and materials

All data generated or analysed during this study are included in this published article.

Abbreviations

CagA:

Cytotoxin-associated gene A

CEA:

Carcinoembryonic antigen

DLBCL:

Diffuse large B-cell lymphoma

ECOG:

Eastern Cooperative Oncology Group

EFS:

Progression-free survival

GCB:

Germinal center B-cell

H. pylori :

Helicobacter pylori

IPI:

International Prognostic Index

LDH:

Lactate dehydrogenase

MALT:

Mucosa-associated lymphoid tissue

NHL:

Non-Hodgkin’s Lymphomas

OS:

Overall survival

References

  1. Sabattini E, Bacci F, Sagramoso C, et al. WHO classification of tumours of haematopoietic and lymphoid tissues in 2008: an overview. Pathologica. 2010;102:83–7.

    CAS  PubMed  Google Scholar 

  2. Tondini C, Giardini R, Bozzetti F, et al. Combined modality treatment for primary gastrointestinal non-Hodgkin’s lymphoma: the Milan Cancer institute experience. Ann Oncol. 1993;4:831–7.

    Article  CAS  PubMed  Google Scholar 

  3. Howell JM, Auer-Grzesiak I, Zhang J, et al. Increasing incidence rates, distribution and histological characteristics of primary gastrointestinal non-Hodgkin lymphoma in a North American population. Can J Gastroenterol. 2012;26:452–6.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Jaffe ES, Harris NL, Stein H, et al. Classification of lymphoid neoplasms: the microscope as a tool for disease discovery. Blood. 2008;112:4384–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Psyrri A, Papageorgiou S, Economopoulos T. Primary extranodal lymphomas of stomach: clinical presentation, diagnostic pitfalls and management. Ann Oncol. 2008;19:1992–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hatakeyama M. Oncogenic mechanisms of the helicobacter pylori CagA protein. Nat Rev Cancer. 2004;4:688–94.

    Article  CAS  PubMed  Google Scholar 

  7. Schistosomes, liver flukes and Helicobacter pylori. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Lyon, 7–14 June 1994. IARC Monogr Eval Carcinog Risks Hum. 1994;61:1–241.

    Google Scholar 

  8. Wotherspoon AC, Ortiz-Hidalgo C, Falzon MR, et al. Helicobacter pylori-associated gastritis and primary B-cell gastric lymphoma. Lancet. 1991;338:1175–6.

    Article  CAS  PubMed  Google Scholar 

  9. Amieva M, Peek RM. Pathobiology of helicobacter pylori-induced gastric Cancer. Gastroenterology. 2016;150:64–78.

    Article  CAS  PubMed  Google Scholar 

  10. Suzuki T, Matsuo K, Ito H, et al. A past history of gastric ulcers and helicobacter pylori infection increase the risk of gastric malignant lymphoma. Carcinogenesis. 2006;27:1391–7.

    Article  CAS  PubMed  Google Scholar 

  11. Meimarakis G, Winter H, Assmann I, et al. Helicobacter pylori as a prognostic indicator after curative resection of gastric carcinoma: a prospective study. Lancet Oncol. 2006;7:211–22.

    Article  PubMed  Google Scholar 

  12. Marrelli D, Pedrazzani C, Berardi A, et al. Negative helicobacter pylori status is associated with poor prognosis in patients with gastric cancer. Cancer. 2009;115:2071–80.

    Article  CAS  PubMed  Google Scholar 

  13. Postlewait LM, Squires MH, Kooby DA, et al. Preoperative helicobacter pylori infection is associated with increased survival after resection of gastric adenocarcinoma. Ann Surg Oncol. 2016;23:1225–33.

    Article  PubMed  Google Scholar 

  14. Hussell T, Isaacson PG, Crabtree JE, et al. The response of cells from low-grade B-cell gastric lymphomas of mucosa-associated lymphoid tissue to helicobacter pylori. Lancet. 1993;342:571–4.

    Article  CAS  PubMed  Google Scholar 

  15. Neubauer A, Thiede C, Morgner A, et al. Cure of helicobacter pylori infection and duration of remission of low-grade gastric mucosa-associated lymphoid tissue lymphoma. J Natl Cancer Inst. 1997;89:1350–5.

    Article  CAS  PubMed  Google Scholar 

  16. Swerdlow SH, Campo E, Harris NL, et al. WHO classification of tumors of hematopoietic and lymphoid tissues. 4th ed. Lyon: IARC Press; 2008.

    Google Scholar 

  17. Chen LT, Lin JT, Shyu RY, et al. Prospective study of helicobacter pylori eradication therapy in stage I(E) high-grade mucosa-associated lymphoid tissue lymphoma of the stomach. J Clin Oncol. 2001;19:4245–51.

    Article  CAS  PubMed  Google Scholar 

  18. Morgner A, Miehlke S, Fischbach W, et al. Complete remission of primary high-grade B-cell gastric lymphoma after cure of helicobacter pylori infection. J Clin Oncol. 2001;19:2041–8.

    Article  CAS  PubMed  Google Scholar 

  19. Kuo SH, Yeh KH, Chen LT, et al. Helicobacter pylori-related diffuse large B-cell lymphoma of the stomach: a distinct entity with lower aggressiveness and higher chemosensitivity. Blood Cancer J. 2014;4:e220.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Huang WT, Kuo SH, Cheng AL, et al. Inhibition of ZEB1 by miR-200 characterizes helicobacter pylori-positive gastric diffuse large B-cell lymphoma with a less aggressive behavior. Mod Pathol. 2014;27:1116–25.

    Article  CAS  PubMed  Google Scholar 

  21. Chan JK, Ng CS, Isaacson PG. Relationship between high-grade lymphoma and low-grade B-cell mucosa-associated lymphoid tissue lymphoma (MALToma) of the stomach. Am J Pathol. 1990;136:1153–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. de Jong D, Boot H, van Heerde P, et al. Histological grading in gastric lymphoma: pretreatment criteria and clinical relevance. Gastroenterology. 1997;112:1466–74.

    Article  PubMed  Google Scholar 

  23. Rohatiner A, d'Amore F, Coiffier B, et al. Report on a workshop convened to discuss the pathological and staging classifications of gastrointestinal tract lymphoma. Ann Oncol. 1994;5:397–400.

    Article  CAS  PubMed  Google Scholar 

  24. Nakamura S, Sugiyama T, Matsumoto T, et al. Long-term clinical outcome of gastric MALT lymphoma after eradication of helicobacter pylori: a multicentre cohort follow-up study of 420 patients in Japan. Gut. 2012;61:507–13.

    Article  PubMed  Google Scholar 

  25. Fischbach W, Goebeler ME, Ruskone-Fourmestraux A, et al. Most patients with minimal histological residuals of gastric MALT lymphoma after successful eradication of helicobacter pylori can be managed safely by a watch and wait strategy: experience from a large international series. Gut. 2007;56:1685–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chen LT, Lin JT, Tai JJ, et al. Long-term results of anti-helicobacter pylori therapy in early-stage gastric high-grade transformed MALT lymphoma. J Natl Cancer Inst. 2005;97:1345–53.

    Article  CAS  PubMed  Google Scholar 

  27. Sugimoto M, Kajimura M, Sato Y, et al. Regression of primary gastric diffuse large B-cell lymphoma after eradication of helicobacter pylori. Gastrointest Endosc. 2001;54:643–5.

    Article  CAS  PubMed  Google Scholar 

  28. Alsolaiman MM, Bakis G, Nazeer T, et al. Five years of complete remission of gastric diffuse large B cell lymphoma after eradication of helicobacter pylori infection. Gut. 2003;52:507–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kuo SH, Yeh KH, Wu MS, et al. Helicobacter pylori eradication therapy is effective in the treatment of early-stage H pylori-positive gastric diffuse large B-cell lymphomas. Blood. 2012;119:4838–44 quiz 5057.

    Article  CAS  PubMed  Google Scholar 

  30. Murata-Kamiya N, Kikuchi K, Hayashi T, et al. Helicobacter pylori exploits host membrane phosphatidylserine for delivery, localization, and pathophysiological action of the CagA oncoprotein. Cell Host Microbe. 2010;7:399–411.

    Article  CAS  PubMed  Google Scholar 

  31. Negrini R, Savio A, Poiesi C, et al. Antigenic mimicry between helicobacter pylori and gastric mucosa in the pathogenesis of body atrophic gastritis. Gastroenterology. 1996;111:655–65.

    Article  CAS  PubMed  Google Scholar 

  32. Xue LJ, Su QS, Yang JH, et al. Autoimmune responses induced by helicobacter pylori improve the prognosis of gastric carcinoma. Med Hypotheses. 2008;70:273–6.

    Article  CAS  PubMed  Google Scholar 

  33. Kuo SH, Chen LT, Lin CW, et al. Detection of the helicobacter pylori CagA protein in gastric mucosa-associated lymphoid tissue lymphoma cells: clinical and biological significance. Blood Cancer J. 2013;3:e125.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Lemma S, Karihtala P, Haapasaari KM, et al. Biological roles and prognostic values of the epithelial-mesenchymal transition-mediating transcription factors twist, ZEB1 and slug in diffuse large B-cell lymphoma. Histopathology. 2013;62:326–33.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

Study concept and design: XLM; Acquisition of data: YC, JZ and YL; Analysis and interpretation of data: YC and XLM; Drafting of the manuscript: YC and YNX; Critical revision of the manuscript for important intellectual content: RF Z, YNX, and XLM; Statistical analysis: YC, YL and JZ; Study supervision: XLM. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Xuelei Ma.

Ethics declarations

Ethics approval and consent to participate

Approval for this retrospective research was obtained from the institutional review board of West China hospital, Sichuan University. Written informed consent was obtained from all included patients.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cheng, Y., Xiao, Y., Zhou, R. et al. Prognostic significance of helicobacter pylori-infection in gastric diffuse large B-cell lymphoma. BMC Cancer 19, 842 (2019). https://doi.org/10.1186/s12885-019-6067-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-019-6067-5

Keywords