Mucosa associated lymphoid tissue malt b-cell lymphomas là gì

Extranodal marginal zone (MZ) B-cell lymphomas of the mucosa-associated lymphoid tissue (MALT) arise from lymphoid populations that are induced by chronic inflammation in extranodal sites. The best evidence of an etiopathogenetic link is provided by the association between Helicobacter pylori–positive gastritis and gastric MALT lymphoma. Indeed, successful eradication of this microorganism with antibiotics can be followed by gastric MALT lymphoma regression in most cases. Other microbial agents have been implicated in the pathogenesis of MZ lymphoma arising at different sites. Apart from gastric MALT lymphoma, antibiotic therapies have been adequately tested only in ocular adnexal MALT lymphomas where upfront doxycycline may be a reasonable and effective initial treatment of patients with Chlamydophila psittaci–positive lymphoma before considering more aggressive strategies. In all other instances, antibiotic treatment of nongastric lymphomas remains investigational. Indeed, there is no clear consensus for the treatment of patients with gastric MALT lymphoma requiring further treatment beyond H pylori eradication or with extensive disease. Both radiotherapy and systemic treatments with chemotherapy and anti-CD20 antibodies are efficacious and thus the experience of individual centers and each patient’s preferences in terms of adverse effects are important parameters in the decision process.

The peculiar biology of MALT lymphomas

Marginal zone (MZ) lymphomas (MZLs) represent a group of lymphomas originating from B lymphocytes of the “marginal zone,” which is the external part of the secondary lymphoid follicles. This anatomic compartment is more prominent in lymphoid organs that are continuously exposed to not only a high flow of external antigens, particularly the spleen, but also the mucosa-associated lymphoid tissue (MALT) and the mesenteric lymph nodes. The B cells resident in the MZ function as innate-like lymphocytes that mount rapid antibody responses to both T-cell–dependent and –independent antigens.1 Most of the MZ lymphocytes are B cells that are involved in the T-cell–independent early immune response and express a restricted immunoglobulin repertoire. Post-germinal center memory B cells, needed for the T-cell–dependent immune response, are also localized in the MZ, as well as a variety of other immune system cells (plasma cells, dendritic cells, macrophages, T cells, and granulocytes) that interact with circulating antigens.

In the World Health Organization (WHO) classification, there are 3 different MZL entities with specific diagnostic criteria, different behavior, and therapeutic implications: the extranodal MZL of MALT type (MALT lymphoma), the splenic MZL, with or without villous lymphocytes, and nodal MZL.2 The genetic relationship between the 3 MZL subtypes (nodal, extranodal, and splenic) is still unclear. A comprehensive analysis of DNA copy-number changes in a very large series of 218 MZL patients showed that the 3 MZL types share recurrent trisomies of chromosomes 3 and 18 and deletions at 6q23 (TNFAIP3).3 MALT lymphoma presents more frequently gains at 3p, 6p, 18p, and the del(6q23).3 Different from the other 2 MZL types, MALT lymphoma presents recurrent chromosomal translocations4-11 (Table 1), and at least 3 of them lead to the activation of the nuclear factor κB (NF-κB) pathway,23,24 which can also be constitutively turned on due to the inactivation of TNFAIP3 by either somatic mutation and/or del(6q23)3,25,26 or, possibly, by stimulation of the Toll-like receptor signaling as suggested in splenic MZLs.27 Nodal and splenic MZLs share recurrent mutations affecting the Notch pathway and the transcription factor KLF2, but differ for the inactivation of 2 tumor-suppressor genes, detected exclusively (PTPRD) or much more commonly (KMT2D/MLL2) in the nodal type.28,29

Table 1

Most common genetic aberrations detected in MALT lymphomas

Genetic lesion3-22 Involved genesDeregulated pathwayPrevalence, %Anatomical sitesClinical relevancet(11;18)(q21;q21) BIRC3-MALT1 NF-κB 15-40 Stomach, lung Antibiotic resistance alkylating agents resistance? t(14;18)(q32;q21) IGHV-MALT1 NF-κB 20 Lung, salivary gland, skin, ocular adnexa Antibiotic resistance? t(1;14)(p22;q32) IGHV-BCL10 NF-κB <5 Stomach, lung Antibiotic resistance t(3;14)(p13;q32) IGHV-FOXP1 Wnt* <5 Unclear Transformation risk? t(9;14)(p24;q32) IGHV-JMJD2C Chromatin remodeling† <5 Unclear t(X;14)(p11;q32) IGHV-GPR34 NF-κB ? <5 Unclear t(5;14)(q34;q32) IGHV- TENM2 Unclear <5 Unclear Trisomy 3 Unclear Unclear 20-40 Equal distribution Inferior outcome? Trisomy 18 Unclear Unclear 20-40 Equal distribution del(6q23) TNFAIP3 NF-κB 15-30 Equal distribution

Genetic lesion3-22 Involved genesDeregulated pathwayPrevalence, %Anatomical sitesClinical relevancet(11;18)(q21;q21) BIRC3-MALT1 NF-κB 15-40 Stomach, lung Antibiotic resistance alkylating agents resistance? t(14;18)(q32;q21) IGHV-MALT1 NF-κB 20 Lung, salivary gland, skin, ocular adnexa Antibiotic resistance? t(1;14)(p22;q32) IGHV-BCL10 NF-κB <5 Stomach, lung Antibiotic resistance t(3;14)(p13;q32) IGHV-FOXP1 Wnt* <5 Unclear Transformation risk? t(9;14)(p24;q32) IGHV-JMJD2C Chromatin remodeling† <5 Unclear t(X;14)(p11;q32) IGHV-GPR34 NF-κB ? <5 Unclear t(5;14)(q34;q32) IGHV- TENM2 Unclear <5 Unclear Trisomy 3 Unclear Unclear 20-40 Equal distribution Inferior outcome? Trisomy 18 Unclear Unclear 20-40 Equal distribution del(6q23) TNFAIP3 NF-κB 15-30 Equal distribution

*

DLBCLs bearing the same chromosomal translocation show deregulated Wnt signaling.21

JMJD2C is amplified and overexpressed in other lymphoma subtypes in which its genetic silencing leads to decreased levels of H3K9me3, a marker of transcriptional repression.22

MALT lymphoma is the commonest MZL type, accounting for 5% to 8% of all B-cell lymphomas,30,31 and has been described in virtually all tissues, often in organs that are normally devoid of germinal centers. Indeed, they arise from lymphoid populations that are induced by chronic inflammation in extranodal sites. The most frequently affected organ is the stomach, where MALT lymphoma has been incontrovertibly associated with the chronic gastritis induced by Helicobacter pylori whereas a possible etiologic link has been shown between other microorganisms and MALT lymphomas arising in other anatomical sites.32 In addition to infections, chronic inflammation caused by autoimmune diseases, such as Sjögren syndrome or Hashimoto thyroiditis, can also carry a significant risk for the development of MALT lymphoma.

Besides the continuous antigenic stimulation, additional oncogenic events play a relevant role in lymphoma growth and progression to the point where the lymphoproliferative process becomes frankly malignant and, eventually, independent of the antigenic drive.32 This makes the differential diagnosis between the preexisting chronic inflammation and the MALT lymphoma not always straightforward: clonal B-cell expansions can be detected in benign inflammatory tissues, particularly in the context of autoimmune reactions. Also, the presence of the typical lymphoepithelial lesions is neither essential nor pathognomonic for the diagnosis of MALT lymphoma, as they can be detected in some reactive conditions or in other lymphoma subtypes.33 Hence, having the diagnosis confirmed by an expert hematopathologist to avoid overtreatment of benign conditions is recommended.33

Diverse pathogenetic mechanisms may lead to diverse clinical outcomes not only from organ to organ31 but also within the same organ34 ; these differences, particularly with respect to personalized medicine, might impact therapeutic approaches. This review will summarize the many faces of MALT lymphoma pathogenesis and the current evidence for site-directed treatments.

Antigen drive and genetic lesions

MZ B cells are continuously exposed to exogenous antigens and have a physiologically reduced threshold for triggering proliferation, which may predispose them to malignant transformation.1

As stated in the previous section, extranodal MZL most frequently occur in organs normally devoid of germinal centers following a process of chronic inflammation and antigenic stimulation, where genes that regulate apoptosis, cell survival, and proliferation play a prominent role. Autoimmune disorders are, in this context, considered a potential risk factor for the development of lymphomas. Indeed, patients with Sjögren syndrome have an extremely increased risk of developing a MZL.35-38 The mechanisms might be, however, distinct in each autoimmune disease. In the case of the Sjögren syndrome, it has been hypothesized that a local chronic antigen drive activates the development of organized lymphoid tissue in lacrimal and salivary glands and that CD40/CD40 ligand (CD40L) and BCL2 family proteins together with the overexpression of B-cell–activating factor (BAFF) may lead to excessive autoantibody production and reduced apoptosis, providing a stimulus for sustained proliferation of B cells.37,39,40 Individual genetic differences, highlighted by the recent report of polymorphisms near the BTNL2 and HLA-B genes in the HLA region,41 influence the susceptibility to develop MZL.

MALT lymphoma presents somatically mutated immunoglobulin heavy chain variable region (IGHV) genes in nearly all cases. IGHV sequence analysis shows a pattern of somatic hypermutation and rearrangement, suggesting that tumor cells have undergone antigen selection in germinal centers.42 The presence of the so-called ongoing mutations (intraclonal variation) and the biased usage of some IGHV segments indicate that the expansion of lymphoma cells could still be antigen-driven. Interestingly, a specific usage of different restricted IGHV families appears associated with different anatomical sites or with particular clinical and genetic features: IGHVH1-69 in salivary gland lymphomas; IGHVH3-30 or IGHVH3-23 in gastric MALT lymphomas responsive to H pylori eradication and without the t(11;18) translocation; IGHVH4-34 in orbital adnexal lymphomas; IGHV3 and IGHV4 families in pulmonary lymphomas; and IGHVH1-69 or IGHVH4-59 in cutaneous lymphomas.43 Also, the antibodies expressed by MALT lymphoma cells can present, although not always, specificity for self-antigens.44-46

As a clinicopathological entity, MALT lymphomas from different anatomical sites share common histological, clinical, and genetic features, but differences do exist.47 The autoimmune or infective disorders that precede the lymphoma differ from site to site, and this can impact the clinical features, and, possibly, the genetic identity of the lymphoma. Indeed, the recurrent chromosomal aberrations occur at frequencies that vary according to anatomical localization.9,12,48

Main clinical characteristics of MALT lymphoma

The stomach is the commonest localization; frequent nongastric sites are: salivary glands, skin, orbits and conjunctiva, lung, thyroid, upper airways, breast, other gastrointestinal (GI) sites, and liver.47,49-51 The anatomic site may have prognostic relevance because of organ-specific clinical problems but, because different genetic lesions may be associated with different localizations,12 it is possible that the different sites have a distinct natural history. In a study evaluating the long-term outcome of 167 patients with localized (stage IE and IIE) MALT lymphoma treated with involved field radiotherapy, gastric and thyroid lymphomas had a significantly better outcome and distant failures were more common for other sites.52 In general, despite frequent relapses, MALT lymphomas most often maintain an indolent course.47 In the above-mentioned study, the 10-year recurrence-free rate was 76%, the overall survival rate was 87%, and the cause-specific survival rate was 98%.52 Similar results were reported in a survey of 490 patients with stage I-II MALT lymphoma treated with radiotherapy only; the 10-year overall and recurrence-free survival were 79% and 57%, respectively, and patients with stomach or head and neck lymphomas had longer relapse-free survival.53

Within the same organ, the outcome may be different, possibly as a result of different pathogenetic pathways as suggested by the finding that, in gastric MALT lymphoma, the presence of MALT1 translocation confers resistance to antibiotic treatment or that, among the patients with salivary gland lymphomas, those with a history of Sjögren syndrome have a better survival compared with those without.34 Because lymphomagenesis in these patients is related to chronic immune stimulation and dysregulation, these outcome differences may be reflective of the differing biology of MALT development. The prognostic impact of concomitant autoimmune disease remains, however, not yet fully elucidated. In a series of 158 MALT lymphoma patients, those with autoimmune disease were predominantly women and significantly younger at lymphoma diagnosis (56 vs 67 years), with a significantly higher rate of extragastric lymphomas.54 The clinical course, however, did not appear to be adversely influenced by the presence of autoimmune diseases; apart from a lower response rate to H pylori eradication in patients with gastric lymphoma, neither times to relapse or survival significantly differed.54

Although up to one-third of diffuse large B-cell lymphoma (DLBCL) arise from extranodal sites, histological transformation of MALT lymphoma to large-cell lymphoma is comparatively less frequent than for follicular lymphomas with an occurrence well below 10% in most series, also occurring as a late event, independent from dissemination.13,51,52,55-57

MALT lymphoma characteristically remains localized for a prolonged period within the tissue of origin, but involvement of regional lymph nodes and spreading to multiple sites may occur. Localized MALT lymphoma is often multifocal within the involved organ (ie, stomach, skin), although this may not reflect a truly disseminated disease. The latter is reported in up to one-quarter of cases and is more common in non-GI MALT lymphomas.47,50,51,55 Bone marrow infiltration is observed at a similar frequency, occurring in up to 20% of cases.58 Patients with lymph node or bone marrow involvement at presentation, but not those with involvement of multiple mucosal sites, are associated with a worse prognosis.47

Due to the risk of occult-disseminated disease, extensive initial staging assessment is indicated regardless of the presentation site,59 particularly if antibiotic treatment or localized radiotherapy is planned. Besides standard computerized tomography (CT) scan imaging of the chest and abdomen, recommended site-specific procedures are reported in Table 2.33,59 The value of the positron emission tomography (PET) scan is still unclear. In general, the use of PET-CT scan in the routine staging of MZL is not recommended,33,59 except for selected cases (ie, when a transformation to high-grade lymphoma is suspected). However, there is some growing evidence that many nongastric sites are usually PET-positive.60,61 In a meta-analysis of the published literature up to 2014,60 the pooled detection rate of 18fluorodeoxyglucose (F-FDG) PET or PET-CT in MALT lymphomas was 71%, and appeared particularly high in the pulmonary (94%) and head and neck (90%) localizations, showing that MALT lymphoma can often be FDG-avid and suggesting a potentially relevant role of PET-CT in the initial evaluation of these patients, especially when the disease is apparently localized and radiotherapy is planned.

Table 2

Recommended site-specific workup in MALT lymphomas

MALT lymphoma siteSite-specific staging proceduresStomach Ear/nose/throat examination, EGD, endoscopic ultrasound to evaluate regional lymph nodes and gastric wall infiltration, search for H pylori (histochemistry, serology, breath test, fecal antigen), search for MALT1 translocation by FISH Salivary glands Ear/nose/throat examination and ultrasound. Anti-SSA or anti-SSB antibodies for possible association with Sjögren syndrome Thyroid Ultrasound ± CT scan of the neck and thyroid function tests Lung Bronchoscopy with bronchoalveolar lavage Small intestine Search for C jejuni in the tumor biopsy (PCR, immunohistochemistry or in situ hybridization) Large intestine Colonoscopy Breast Mammography and MRI Ocular adnexa MRI and ophthalmologic examination. Search for C psittaci in the tumor biopsy and blood mononuclear cells by PCR may be considered Skin Search for B burgdorferi in the tumor biopsy by PCR may be considered in areas where it is endemic

MALT lymphoma siteSite-specific staging proceduresStomach Ear/nose/throat examination, EGD, endoscopic ultrasound to evaluate regional lymph nodes and gastric wall infiltration, search for H pylori (histochemistry, serology, breath test, fecal antigen), search for MALT1 translocation by FISH Salivary glands Ear/nose/throat examination and ultrasound. Anti-SSA or anti-SSB antibodies for possible association with Sjögren syndrome Thyroid Ultrasound ± CT scan of the neck and thyroid function tests Lung Bronchoscopy with bronchoalveolar lavage Small intestine Search for C jejuni in the tumor biopsy (PCR, immunohistochemistry or in situ hybridization) Large intestine Colonoscopy Breast Mammography and MRI Ocular adnexa MRI and ophthalmologic examination. Search for C psittaci in the tumor biopsy and blood mononuclear cells by PCR may be considered Skin Search for B burgdorferi in the tumor biopsy by PCR may be considered in areas where it is endemic

EGD, esophagogastroduodenoscopy; FISH, fluorescence in situ hybridization; MRI, magnetic resonance imaging; PCR, polymerase chain reaction; SSA, Sjögren syndrome A; SSB, Sjögren syndrome B.

Association of different infectious agents with MALT lymphomas at various anatomical sites: therapeutic implications

Several lines of epidemiologic, biologic, and clinical evidence indicate that gastric MALT lymphoma arises from MALT acquired as a consequence of chronic H pylori infection. Outside of the stomach, the acquisition of MALT can be induced by a series of agents, which are different for each anatomic site. Other bacterial infections have been implicated in the pathogenesis of MZL arising in the skin (Borrelia burgdorferi),62 in the ocular adnexa (Chlamydophila psittaci),63 in the small intestine (Campylobacter jejuni),64 and possibly in the lung (Achromobacter xylosoxidans).65 An increased risk has been reported in patients with chronic hepatitis C virus (HCV) infection to develop not only splenic and nodal MZLs but also MALT lymphomas.36,66,67 The association with HCV, however, shows considerable and not entirely explained geographic discrepancies.68 These site-specific biological differences might influence outcome, and recognition of the driving source of the antigenic stimulation in different tissues may have important therapeutic implications. Although antibiotic therapy is nowadays well established for primary gastric MALT lymphoma, much less is known about the value of anti-infectious therapy in other MALT lymphomas (Table 3 69-72 ).

Table 3

Antibiotic-induced lymphoma remission in MALT lymphomas

Involved organTargeted pathogenAntibiotic regimenNo. of patientsType of studyOverall lymphoma remission rate, %Stomach H pylori Mostly PPI plus clarithromycin-based triple therapy with either amoxicillin or metronidazole for 10-14 d 1408 32 studies either retrospective or prospective 77.5 Ocular adnexa C psittaci Doxycycline 100 mg 120 2 prospective, 48 Twice daily ×21 d 4 retrospective, 1 case report Clarithromycin* 500 mg 11 Prospective 45 Twice daily ×6 mo Clarithromycin* 2 g/d, days 1-14, every 21 d (4 courses) 23 Prospective 52 Skin B burgdorferi Ceftriaxone 2 g/d ×14 d (in most cases) 5 Case reports 40

Involved organTargeted pathogenAntibiotic regimenNo. of patientsType of studyOverall lymphoma remission rate, %Stomach H pylori Mostly PPI plus clarithromycin-based triple therapy with either amoxicillin or metronidazole for 10-14 d 1408 32 studies either retrospective or prospective 77.5 Ocular adnexa C psittaci Doxycycline 100 mg 120 2 prospective, 48 Twice daily ×21 d 4 retrospective, 1 case report Clarithromycin* 500 mg 11 Prospective 45 Twice daily ×6 mo Clarithromycin* 2 g/d, days 1-14, every 21 d (4 courses) 23 Prospective 52 Skin B burgdorferi Ceftriaxone 2 g/d ×14 d (in most cases) 5 Case reports 40

*

The clarithromycin activity may also depend on the immunomodulatory and direct antitumor effect of this macrolide antibiotic.71

Helicobacter pylori and the gastric MALT lymphoma pathogenetic model

Initially, H pylori was demonstrated in the gastric mucosa of over 90% of gastric MALT lymphoma cases,73 but there are both geographical74 and temporal variations.75 In particular, a population-based study from Northern Italy showed a declining incidence of H pylori–associated gastric MALT lymphomas in the last decade, most likely due not only to a decreasing prevalence of the infection but also to the now common policy of an early generalized use of proton pump inhibitors (PPIs) without a diagnostic gastroscopy in patients with acid peptic disease symptoms.75

Direct evidence confirming the importance of H pylori in the pathogenesis of gastric lymphoma derives from studies detecting the lymphoma B-cell clone in the chronic gastritis that preceded the lymphoma67,76 and from a series of preclinical studies showing that H pylori can contribute to MALT lymphoma pathogenesis both directly, acting on the still normal and then transformed B cells, and indirectly, affecting other immune cells such as T cells.32,77 A main role is played by the H pylori cytotoxin-associated gene A (CagA) protein, also involved in gastric cancer pathogenesis.78,79 Interindividual differences in antioxidative capacity and in the cellular inflammatory responses to H pylori may represent the genetic background of the H pylori–associated gastric lymphomagenesis.80,81

All of the above-summarized findings are in keeping with a possible model of multistage development and progression from chronic gastritis to gastric lymphoma that starts with chronic H pylori infection, stimulating the formation of a lymphocytic infiltration in the gastric mucosa. As a result of an antigenic stimulation (autoantigens and T cells specific for H pylori) combined with a direct effect on B cells, the latter proliferate and may occasionally undergo neoplastic transformation following the acquisition of genetic abnormalities, perhaps facilitated by the presence of free radicals.10,80 The accumulation of genetic abnormalities would be associated with both a loss of dependency from antigenic stimulation (with subsequent antibiotic resistance) as well as a possible histological transformation. Notably, although additional evidence derived from large prospective studies is needed before routinely adopting such an approach, pathological lymphoma remissions after first-line H pylori eradication therapy have also been reported in some patients with H pylori–positive early-stage DLBCL of the stomach with or without concomitant or prior histological evidence of MALT lymphoma.82-84 This finding suggests that the loss of antigen dependence and high-grade transformation may be separate events in the progression of gastric lymphoma. Of clinical relevance, although MALT lymphomas bearing the t(11;18) present a lower risk of transformation to DLBCL, the t(11;18)-positive primary gastric MALT lymphomas have a low probability of response to antibiotics and are more commonly H pylori negative, with more advanced disease.14,85,86 Also, the t(3;14) has been associated with a risk of transformation to high-grade tumors.13,15

Antibiotic treatment of Helicobacter pylori–positive gastric MALT lymphoma

Eradication of H pylori with antibiotics should be the sole initial therapy for a localized H pylori–positive gastric MALT lymphoma (Figure 1). It results in lymphoma regression and long-term clinical control in most patients.69,87 Several effective anti-H pylori treatments are available. The choice should be based on the epidemiology of the infection and on the expected antibiotic resistance in different countries. The most commonly used regimen comprises a PPI associated with clarithromycin and amoxicillin, administered for 10 to 14 days. Metronidazole can be used instead of amoxicillin for penicillin-allergic patients. H pylori eradication with antibiotics leads to gastric MALT lymphoma regression in 75% of cases.69

Figure 1

Mucosa associated lymphoid tissue malt b-cell lymphomas là gì

Treatment algorithm for the management of gastric MALT lymphoma confined to the stomach, with or without regional lymph node involvement. It is currently recommended that gastric biopsies are evaluated using the Group d’Etude des Lymphomes de l’Adult (GELA) scoring system.33 *Indications to treat comprise overt progression, deep gastric wall invasion, regional lymph node involvement, t(11;18) translocation. EGD, esophagogastroduodenoscopy; RT, involved-field radiotherapy (24-30 Gy to the stomach and perigastric nodes given in 3-4 weeks).

Figure 1

Mucosa associated lymphoid tissue malt b-cell lymphomas là gì

Treatment algorithm for the management of gastric MALT lymphoma confined to the stomach, with or without regional lymph node involvement. It is currently recommended that gastric biopsies are evaluated using the Group d’Etude des Lymphomes de l’Adult (GELA) scoring system.33 *Indications to treat comprise overt progression, deep gastric wall invasion, regional lymph node involvement, t(11;18) translocation. EGD, esophagogastroduodenoscopy; RT, involved-field radiotherapy (24-30 Gy to the stomach and perigastric nodes given in 3-4 weeks).

Close modal

Detailed guidelines for response assessment and follow-up have been recently published33 and our current algorithm for the management of stage IE-IIE gastric MALT lymphoma is summarized in Figure 1. It is important to recall that transient histological relapses can be observed in endoscopic biopsies during long-term follow-up, but they tend to be self-limiting, and importantly without the stimulus from H pylori reinfection, they do not implicate a true clinical relapse. Hence, when persistent but not progressive residual disease or histological relapse is documented, a “wait-and-see” policy seems safe.87-90 Nevertheless, a long-term careful endoscopic and systemic follow-up (clinical examination, blood counts, and minimal adequate radiological or ultrasound examinations every 12-18 months) is strongly advisable for all patients. Indeed, the risk of gastric adenocarcinoma among individual with gastric MALT lymphoma can be up to sixfold higher, and the risk of other lymphomas is higher than in the general population.91,92

Antibiotic treatment of Helicobacter pylori–negative gastric MALT lymphoma

There are also reports of lymphoma regression following antibiotics in H pylori–negative patients, possibly due to a false-negative test or to infection by other Helicobacter species.93 Hence, first-line therapy with antibiotics may be considered at least in those patients without the t(11;18) translocation. However, an oncological treatment is to be considered when no signs of lymphoma regression are seen at a repeated endoscopy assessment 2 to 3 months after antibiotics administration.

Chlamydophila psittaci and ocular adnexal MZL

Besides H pylori, C psittaci is the most thoroughly studied among the other bacteria reported to have a potential pathogenetic role in MZL. Chlamydophila is the etiologic agent of psittacosis, an infection caused by exposure to infected animals. The presence of C psittaci DNA has been detected not only in a variable percentage of MZL, mainly of the ocular adnexae (ie, conjunctiva, lachrymal gland, orbital fat, eyelid, lachrymal sac), but also in MZL of the lungs, skin, thyroid gland, and salivary glands. However, it should be noted that the prevalence of C psittaci infection in ocular adnexal marginal zone lymphoma (OAMZL) varies among countries and different regions within the same country, being higher in Italy, Austria, Korea, and Germany (with prevalence rates up to 80%), and virtually absent in Japan, France, and China.32

Evidence supporting a pathogenic association between C psittaci and the development of MALT lymphoma of the ocular adnexa comprises the identification of Chlamydophilae in tumor tissue by immunohistochemistry and the detection of bacterial DNA in the tumor biopsies, bacterial visualization within tumor-infiltrating macrophages by electronic microscopy, their isolation from conjunctival swabs and from the ocular adnexa peripheral blood of patients,94,95 as well as the description of metachronous C psittaci–related lymphomas observed in the same patient after prolonged exposure to an infected animal.96

Globally, doxycycline has been tested in >100 patients with OAMZL (Table 3), showing an overall response rate (ORR) of around 50%.70 The median time for response after antibiotic therapy is 6 months but in some patients responses are slow and may require up to 36 months.97 In a prospective phase 2 study, C psittaci DNA was detected in nearly 90% of lymphoma biopsy specimens.94 Front-line doxycycline induced Chlamydophila eradication in 14 of 34 patients (48%); 6 patients obtained complete lymphoma regression (ORR, 65%).94 At a median follow-up of 37 months, the 5-year progression-free survival was 55%.94 However, lymphoma regression after doxycycline treatment has been observed in some lymphomas with no C psittaci presence as well as in cases where this treatment failed to eradicate the C psittaci infection,94,97,98 suggesting that other doxycycline-sensitive microorganisms might be involved.99

In a small exploratory study, lymphoma regressions were seen after a 6-month oral clarithromycin regimen in 5 of 11 patients with ocular adnexal MALT lymphoma71 who had been previously unsuccessfully treated with doxycycline. A subsequent phase 2 trial tested the activity of a higher clarithromycin dose in 23 MALT lymphoma patients with relapsed/refractory disease. Ocular adnexae were the most commonly involved organs. Six patients achieved a complete remission (CR) and 6 a partial response (ORR, 52%; 95% confidence interval, 32%-72%). At a median follow-up of 24 months, only 2 patients with responsive disease experienced relapse.72

Borrelia burgdorferi in cutaneous MZL

The prevalence of B burgdorferi infection in patients with cutaneous MZL exhibits important variations among different geographic areas, with higher detection rates in areas where it is endemic. In Europe, DNA of B burgdorferi has been detected in 10% to 42% of patients.99 Anecdotal case reports have shown that the eradication of B burgdorferi following ceftriaxone therapy resulted in regression of an associated cutaneous MZL62,70 (Table 3), corroborating the hypothesis that chronic B burgdorferi infection could represent the background for the development of cutaneous MZL.32

The demonstration of a B burgdorferi infection may be sought in areas of endemicity, where it may have some therapeutic implications; however, the evidence is based on a limited number of patients and therefore no recommendations can be made.

Immunoproliferative small intestinal disease and Campylobacter jejuni

Endemic in the Middle East, the immunoproliferative small intestinal disease (IPSID), previously also known as α-heavy-chain disease or Mediterranean lymphoma, is a special subtype of MALT lymphoma. Sporadic cases can also be diagnosed in Western countries, often among immigrants from the area of endemicity.100,101

IPSID has a long natural history, often over many years, including a potentially reversible early phase. If left untreated, however, the lymphoma can transform to DLBCL.

The restricted geographic distribution of IPSID supports the hypothesis that environmental factors may have a relevant pathogenetic role. It has been known since the 1970s that in its early phases, IPSID can be treated with antibiotic treatment (such as tetracycline or metronidazole and ampicillin for at least 6 months), which can lead to durable remissions in the majority of patients. These results suggest a role for an infectious agent, and Campylobacter jejuni is so far the best, although not necessarily the sole, candidate.64 Indeed, the level of evidence supporting a pathogenetic link of C jejuni with IPSID remains weak, with lymphoma improvement in 2 patients treated with antibiotics against C jejuni99 and a unique study, describing the presence of C jejuni DNA in 5 of 7 archival cases followed by a single confirmatory case report.64

Achromobacter xylosoxidans and pulmonary MALT lymphoma

A xylosoxidans is a gram-negative bacterium characterized by a low virulence but high resistance to antibiotic therapy. It has been recurrently isolated from patients affected by cystic fibrosis and, in these patients, it is correlated with more severe lung damage. A study of 124 pulmonary MALT lymphoma biopsies and 82 nonlymphoma lung biopsies from 6 European countries showed a significantly increased prevalence of A xylosoxidans infection in MALT lymphomas than in control tissues65 Overall, 46% of pulmonary MALT lymphomas and 18% of controls were positive although the infection prevalence among lymphoma patients varied among countries (ranging from 67% in Italy, to 33% in the United Kingdom).65 Further studies are warranted to investigate the potential pathogenetic role of the microorganism because no data demonstrating a causal relationship has yet been provided102 and other microorganisms (Chlamydophila) were reported as possibly involved with MALT lymphoma of the lung.103 Moreover, a previous history of lymphocytic interstitial pneumonia, which is frequently associated with autoimmune disorders, or of other rare nonneoplastic pulmonary lymphoid proliferations (follicular bronchiolitis and nodular lymphoid hyperplasia) support the concept that lymphoma may also evolve from these noninfectious inflammatory processes.104

Other bacterial infections at different MALT sites

Several case reports and small series have described the potential association of various chronic infections with MALT lymphomas localized in the lungs, parotid and salivary glands, breast, thyroid, and bladder but some of these studies showed controversial results.70,99,103 No conclusion can be drawn from the available information on antibiotic treatment in these lymphomas; the published data are scanty and possibly biased by the preferential publication of positive results.70,99

HCV and MALT lymphomas

Numerous epidemiological, clinical, and biological data have suggested an association between HCV infection and the pathogenesis of at least a portion of B-cell lymphomas, including MALT lymphomas, although with important geographical variations. Importantly, the strongest evidence for a causal relationship between HCV and lymphoma comes again from the observation of lymphoma regression after antiviral treatment.68,105,106 Several potential pathogenic mechanisms have been advanced to explain a causative link with lymphoma growth68,107 : a nondirect antigen-driven stimulation; a direct oncogenic role of HCV; a viral immunosuppressive effect on the tumor cells; and the co-infection by another unknown oncogenic virus.

MALT lymphomas in HCV-infected patients are most common at nongastric sites, often the salivary or lacrimal glands.66,108-110 A rare clinical presentation has been described, namely the subcutaneous “lipoma-like” MALT lymphoma. This condition typically affects elderly women and exhibits single or multiple soft and mobile subcutaneous nodules that may regress after HCV eradication.111

Treatment of MALT lymphoma patients with advanced-stage disease or failing antibiotic therapies

There is no clear consensus for the treatment of patients with gastric MALT lymphoma requiring further treatment beyond H pylori eradication or with extensive disease.47,50,51,55

No significant survival difference between patients who received different initial treatments (including chemotherapy alone, surgery alone, surgery with additional chemotherapy, and radiation therapy) has been shown.112,113 However, patients with extragastric lymphoma treated with antibiotics alone may have inferior remission rates and time to next therapy.114 Radiotherapy may be the favored choice for patients with H pylori–negative localized disease or for patients who do not achieve a lymphoma regression following antibiotic therapy.53 Indeed, involved-field radiotherapy to the stomach and perigastric lymph nodes has been shown to allow for excellent disease control, and most reports support the use of a moderate-dose (24-30 Gy given during a period of 3-4 weeks).52,115-117 Literature reports a high rate of local control also in nongastric localizations, in which this therapeutic modality has been recommended by the National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines in Oncology.118 Modern radiotherapy techniques, such as 3-dimensional conformal radiotherapy and intensity-modulated radiotherapy, allow an accurate determination of the clinical target volume, thus reducing toxicity to surrounding organs.116 The moderate radiation doses required for cure (25-35 Gy) are generally associated with mild and reversible acute toxicity and a low risk of long-term side effects, although special caution should be given for specific localizations such as the ocular adnexa or the lung.52,116

In the case of patients with disseminated nongastric MALT lymphoma, observation with careful monitoring can often prove an adequate initial approach. When treatment is required, there is no consensus for the choice of treatment, but rituximab plus chemotherapy appears the most appropriate choice. The treatment approach of disseminated MALT lymphomas is the same in patients with primary gastric and nongastric origin and enrollment in controlled clinical trials is advisable. Indeed, there are no standard recommendations, as only a limited number of drugs and regimens have been specifically tested in MALT lymphomas.119 Oral alkylating agents (either cyclophosphamide or chlorambucil) or purine nucleoside analogs (fludarabine, cladribine) are active as single agents.58,119 Rituximab monotherapy has also been tested in phase 2 studies.120,121 The efficacy and safety of the combination of rituximab plus chlorambucil has been proven in a phase 3 International Extranodal Lymphoma Study Group (IELSG) study in gastric (failing antibiotics) or nongastric MALT lymphomas. In comparison with either rituximab or chlorambucil given as single agent, chlorambucil plus rituximab resulted in significantly superior CR, progression-free and event-free survival rates; however, no overall survival benefit was shown.122,123 The combination of rituximab and bendamustine124 as well as the combination of fludarabine and rituximab have also shown high rates of disease control in smaller nonrandomized studies.125 The significant hematological and infectious toxicity observed with the latter regimen, both during and after therapy, was deemed too high in this patient population.125 As shown in Table 4,126-144 new targeted agents have been poorly studied in MALT lymphomas: only 3 studies127,129,133 were restricted to this entity and included >10 patients.

Table 4

Targeted agents in patients with MALT lymphoma: single agents and combinations

AgentsStudy typeNo. of casesORR, %CR, %Single-agent trials Abexinostat126 Phase 2 13* 15* 0* Bortezomib127 Phase 2 32 48 31 Entospletinib128 Phase 2 17* 18* 0* Everolimus129 Phase 2 16 25 6 Ibrutinib130 Phase 1 4* 25* 0* Idelalisib131,132 Phase 1 3* 0* 0 Idelalisib131,132 Phase 2 9* 56* 11 Lenalidomide133 Phase 2 18 61 33† Tazemetostat134 Phase 1 1* 100* n.a. Thalidomide135 Phase 2 8 0‡ 0‡ Combination trials Bortezomib, rituximab136 Phase 2 8* 50* n.a. Ibrutinib, lenalidomide137 Phase 1 2* 50* 0* Ibrutinib, rituximab, bendamustine138 Phase 1 1* 100* 0* Idelalisib, lenalidomide, rituximab139 Phase 1 1* n.a.§ n.a.§ Lenalidomide, rituximab140 Phase 2 46 80 54 Lenalidomide, rituximab141 Phase 2 27* 89* 67* Lenalidomide, rituximab, bendamustine142 Phase 1 3* 67* 0* Venetoclax, bendamustine, rituximab143 Phase 1 4* 75* 25*

AgentsStudy typeNo. of casesORR, %CR, %Single-agent trials Abexinostat126 Phase 2 13* 15* 0* Bortezomib127 Phase 2 32 48 31 Entospletinib128 Phase 2 17* 18* 0* Everolimus129 Phase 2 16 25 6 Ibrutinib130 Phase 1 4* 25* 0* Idelalisib131,132 Phase 1 3* 0* 0 Idelalisib131,132 Phase 2 9* 56* 11 Lenalidomide133 Phase 2 18 61 33† Tazemetostat134 Phase 1 1* 100* n.a. Thalidomide135 Phase 2 8 0‡ 0‡ Combination trials Bortezomib, rituximab136 Phase 2 8* 50* n.a. Ibrutinib, lenalidomide137 Phase 1 2* 50* 0* Ibrutinib, rituximab, bendamustine138 Phase 1 1* 100* 0* Idelalisib, lenalidomide, rituximab139 Phase 1 1* n.a.§ n.a.§ Lenalidomide, rituximab140 Phase 2 46 80 54 Lenalidomide, rituximab141 Phase 2 27* 89* 67* Lenalidomide, rituximab, bendamustine142 Phase 1 3* 67* 0* Venetoclax, bendamustine, rituximab143 Phase 1 4* 75* 25*

Data were collected from full papers and from abstracts presented at the 2015 meeting of the American Society of Hematology with results available for MZL patients.

n.a., not available.

*

Unspecified whether MALT lymphoma, splenic MZL, or nodal MZL.

39% CR at a later report.144

25% ORR, 25% CR at a later report.144

§

Trial stopped after 7 lymphoma patients due to hepatotoxicity.

Aggressive anthracycline-containing chemotherapy regimens should be reserved for patients with high tumor burden (bulky masses, unfavorable International Prognostic Index) or for those with histological transformation.145

Antibiotic therapy in gastric diffuse large B-cell lymphoma

Although patients with gastric DLBCL can achieve tumor regression after anti-Helicobacter therapy,82-84 additional evidence derived from large prospective studies is needed before routinely adopting this approach, and, at present, we recommend treating gastric large-cell lymphomas according to the guidelines for localized DLBCL.146 Antibiotic therapy as first-line treatment of these patients is not advised outside of clinical trials until evidence is derived from large prospective studies.

Conclusions

No definite guidelines exist for the management of nongastric MALT lymphoma (nor for H pylori–negative cases). Apart from gastric MALT lymphoma, antibiotic therapies have been extensively tested only in ocular adnexal MALT lymphomas where, with negligible toxicity, the outcome of doxycycline therapy, although lacking long-term follow-up information, seems not inferior to the outcome reported for chemotherapy and radiotherapy, suggesting that upfront doxycycline is a reasonable and effective treatment proposal for patients with stage I C psittaci–positive ocular adnexa MALT lymphoma before considering more aggressive strategies.94 In all other instances, antibiotic treatment of nongastric lymphomas remains investigational. Radiotherapy can be effective in providing local disease control even for some patients with disseminated disease.53 However, there is no clear consensus as to whether radiation is more or less effective than systemic therapy in MALT lymphomas at different locations, and the experience of each center and the patient’s preferences in terms of adverse effects are important parameters in the decision process.147 Because no curative treatment exists, expectant observation can be an adequate initial policy in most patients with disseminated disease. In general, the treatment should be ‘‘patient-tailored,’’ taking into account the site, the stage, and the clinical characteristics of the individual patient. When systemic treatment is needed, chemotherapy (and/or immunotherapy with anti-CD20 monoclonal antibodies) can be considered. In this context, enrollment in controlled clinical trials (Table 5) is advisable because only a few compounds and regimens have been specifically tested in the setting of MALT lymphomas.

Table 5

Phase 2 and 3 clinical trials recruiting patients with MALT lymphoma

Study typeClinical trial registration no.Trial armsPatient populationStudy sponsorPhase 3 NCT00877214 Rituximab maintenance of 2 y vs 4 y U Academy Phase 3 NCT01732913 Idelalisib vs idelalisib plus rituximab R/R Industry Phase 3 NCT01938001 Rituximab/lenalidomide vs rituximab R/R Industry Phase 3 NCT01974440 BR or R-CHOP vs BR plus ibrutinib or R-CHOP plus ibrutinib R/R Industry Phase 3 NCT01996865 Lenalidomide/rituximab followed by lenalidomide vs lenalidomide/rituximab followed by rituximab R/R Industry Phase 2 NCT00923663 Lenalidomide R/R Academy Phase 2 NCT01514344 Intralesional rituximab R/R Academy Phase 2 NCT01516606 Clarithromycin R/R Academy Phase 2 NCT01678404 131I-rituximab R/R Academy Phase 2 NCT01808599 Chlorambucil and subcutaneous rituximab U Academy Phase 2 NCT01820910 Doxycycline U Academy Phase 2 NCT02086591 Doxycycline R/R Academy Phase 2 NCT02332980 Pembrolizumab R/R Academy Phase 2 NCT01995669 Lenalidomide plus obinutuzumab R/R Academy Phase 2 NCT02433795 Bendamustine plus rituximab R/R Academy

Study typeClinical trial registration no.Trial armsPatient populationStudy sponsorPhase 3 NCT00877214 Rituximab maintenance of 2 y vs 4 y U Academy Phase 3 NCT01732913 Idelalisib vs idelalisib plus rituximab R/R Industry Phase 3 NCT01938001 Rituximab/lenalidomide vs rituximab R/R Industry Phase 3 NCT01974440 BR or R-CHOP vs BR plus ibrutinib or R-CHOP plus ibrutinib R/R Industry Phase 3 NCT01996865 Lenalidomide/rituximab followed by lenalidomide vs lenalidomide/rituximab followed by rituximab R/R Industry Phase 2 NCT00923663 Lenalidomide R/R Academy Phase 2 NCT01514344 Intralesional rituximab R/R Academy Phase 2 NCT01516606 Clarithromycin R/R Academy Phase 2 NCT01678404 131I-rituximab R/R Academy Phase 2 NCT01808599 Chlorambucil and subcutaneous rituximab U Academy Phase 2 NCT01820910 Doxycycline U Academy Phase 2 NCT02086591 Doxycycline R/R Academy Phase 2 NCT02332980 Pembrolizumab R/R Academy Phase 2 NCT01995669 Lenalidomide plus obinutuzumab R/R Academy Phase 2 NCT02433795 Bendamustine plus rituximab R/R Academy

Trials have been selected if registered at ClinicalTrials.gov, and marked as recruiting on December 12, 2015. Sorted by study type and clinical trial registration number.

BR, bendamustine, rituximab; R-CHOP, rituximab, cyclophosphamide, doxorubicin, vincristin, prednisone; R/R, relapsed/refractory; U, untreated.

Acknowledgments

The authors thank Afua Adjeiwaa Mensah for manuscript review and Rita Gianascio Gianocca for editorial assistance.

Authorship

Contribution: E.Z. and F.B. cowrote the manuscript and approved the final version.

Conflict-of-interest disclosure: E.Z. received advisory honoraria or support of investigator-initiated studies (for the institution) from Celgene, Johnson & Johnson/Janssen, Gilead, Mundipharma, Roche, and Bayer. F.B. received advisory honoraria or research funds (for the institution) from Oncoethix, PIQUR Therapeutics AG, EMD Serono, Bayer AG, Sigma Tau, Italfarmaco, CTI Life Sciences, and Celgene.

Correspondence: Emanuele Zucca, Oncology Institute of Southern Switzerland (IOSI), Ospedale San Giovanni, CH-6500 Bellinzona, Switzerland; e-mail: [email protected].

References

1

, , .

Marginal zone B cells: virtues of innate-like antibody-producing lymphocytes.

,

Nat Rev Immunol

,

2013

, vol.

13

2

(pg.

118

-

132

)

2

, , , et al. ,

WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues

,

2008

Lyon

IARC

3

, , , et al.

Genome-wide DNA profiling of marginal zone lymphomas identifies subtype-specific lesions with an impact on the clinical outcome.

,

Blood

,

2011

, vol.

117

5

(pg.

1595

-

1604

)

4

, , , et al.

The apoptosis inhibitor gene API2 and a novel 18q gene, MLT, are recurrently rearranged in the t(11;18)(q21;q21) associated with mucosa-associated lymphoid tissue lymphomas.

,

Blood

,

1999

, vol.

93

11

(pg.

3601

-

3609

)

5

, , , et al.

Bcl10 is involved in t(1;14)(p22;q32) of MALT B cell lymphoma and mutated in multiple tumor types.

,

Cell

,

1999

, vol.

96

1

(pg.

35

-

45

)

6

, , , et al.

T(14;18)(q32;q21) involving IGH and MALT1 is a frequent chromosomal aberration in MALT lymphoma.

,

Blood

,

2003

, vol.

101

6

(pg.

2335

-

2339

)

7

, , , , .

T(3;14)(p14.1;q32) involving IGH and FOXP1 is a novel recurrent chromosomal aberration in MALT lymphoma.

,

Leukemia

,

2005

, vol.

19

4

(pg.

652

-

658

)

8

, , , et al.

The incidence and anatomic site specificity of chromosomal translocations in primary extranodal marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue (MALT lymphoma) in North America.

,

Am J Surg Pathol

,

2006

, vol.

30

12

(pg.

1546

-

1553

)

9

, , , et al.

Translocations t(11;18)(q21;q21) and t(14;18)(q32;q21) are the main chromosomal abnormalities involving MLT/MALT1 in MALT lymphomas.

,

Leukemia

,

2003

, vol.

17

11

(pg.

2225

-

2229

)

10

, , , et al.

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

, vol.

102

3

(pg.

1012

-

1018

)

11

, , , et al.

FOXP1 abnormalities in lymphoma: translocation breakpoint mapping reveals insights into deregulated transcriptional control.

,

Mod Pathol

,

2008

, vol.

21

7

(pg.

902

-

911

)

12

, , , et al.

Genomic profiles of MALT lymphomas: variability across anatomical sites.

,

Haematologica

,

2011

, vol.

96

7

(pg.

1064

-

1066

)

13

, , , et al.

Forkhead box protein P1 expression in mucosa-associated lymphoid tissue lymphomas predicts poor prognosis and transformation to diffuse large B-cell lymphoma.

,

J Clin Oncol

,

2006

, vol.

24

16

(pg.

2490

-

2497

)

14

, , , et al.

T(11;18) is a marker for all stage gastric MALT lymphomas that will not respond to H. pylori eradication.

,

Gastroenterology

,

2002

, vol.

122

5

(pg.

1286

-

1294

)

15

, , , et al.

Genetic rearrangement of FOXP1 is predominantly detected in a subset of diffuse large B-cell lymphomas with extranodal presentation.

,

Leukemia

,

2006

, vol.

20

7

(pg.

1300

-

1303

)

16

, , , et al.

High incidence of t(11;18)(q21;q21) in Helicobacter pylori-negative gastric MALT lymphoma.

,

Blood

,

2003

, vol.

101

7

(pg.

2547

-

2550

)

17

, , , et al.

Marginal zone B-cell lymphomas of different sites share similar cytogenetic and morphologic features.

,

Blood

,

1996

, vol.

87

1

(pg.

299

-

307

)

18

, , , et al.

t(X;14)(p11.4;q32.33) is recurrent in marginal zone lymphoma and up-regulates GPR34.

,

Haematologica

,

2012

, vol.

97

2

(pg.

184

-

188

)

19

, , , et al.

t(X;14)(p11;q32) in MALT lymphoma involving GPR34 reveals a role for GPR34 in tumor cell growth.

,

Blood

,

2012

, vol.

120

19

(pg.

3949

-

3957

)

20

, , , , , .

Mucosa-associated lymphoid tissue lymphoma: novel translocations including rearrangements of ODZ2, JMJD2C, and CNN3.

,

Clin Cancer Res

,

2008

, vol.

14

20

(pg.

6426

-

6431

)

21

, , , et al.

FOXP1 potentiates Wnt/β-catenin signaling in diffuse large B cell lymphoma.

,

Sci Signal

,

2015

, vol.

8

362

pg.

ra12

22

, , , et al.

Cooperative epigenetic modulation by cancer amplicon genes.

,

Cancer Cell

,

2010

, vol.

18

6

(pg.

590

-

605

)

23

, .

Aberrant NF-kappaB signaling in lymphoma: mechanisms, consequences, and therapeutic implications.

,

Blood

,

2007

, vol.

109

7

(pg.

2700

-

2707

)

24

, , , et al.

Selective expansion of marginal zone B cells in Emicro-API2-MALT1 mice is linked to enhanced IkappaB kinase gamma polyubiquitination.

,

Cancer Res

,

2006

, vol.

66

10

(pg.

5270

-

5277

)

25

, , , et al.

The NF-{kappa}B negative regulator TNFAIP3 (A20) is inactivated by somatic mutations and genomic deletions in marginal zone lymphomas.

,

Blood

,

2009

, vol.

113

20

(pg.

4918

-

4921

)

26

, , , et al.

TNFAIP3/A20 functions as a novel tumor suppressor gene in several subtypes of non-Hodgkin lymphomas.

,

Blood

,

2009

, vol.

114

12

(pg.

2467

-

2475

)

27

, , , et al.

Toll-like receptor stimulation in splenic marginal zone lymphoma can modulate cell signaling, activation and proliferation.

,

Haematologica

,

2015

, vol.

100

11

(pg.

1460

-

1468

)

28

, , , et al.

The coding genome of splenic marginal zone lymphoma: activation of NOTCH2 and other pathways regulating marginal zone development.

,

J Exp Med

,

2012

, vol.

209

9

(pg.

1537

-

1551

)

29

, , , et al.

The coding genome of nodal marginal zone lymphoma reveals recurrent molecular alterations of PTPRD and other Jak/Stat signaling genes [abstract].

,

Blood

,

2014

, vol.

124

21

Abstract 705

30

A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin’s lymphoma. The Non-Hodgkin’s Lymphoma Classification Project.

,

Blood

,

1997

, vol.

89

11

(pg.

3909

-

3918

)

31

, .

Survival of patients with marginal zone lymphoma: analysis of the Surveillance, Epidemiology, and End Results database.

,

Cancer

,

2013

, vol.

119

3

(pg.

629

-

638

)

32

, , , .

Emerging role of infectious etiologies in the pathogenesis of marginal zone B-cell lymphomas.

,

Clin Cancer Res

,

2014

, vol.

20

20

(pg.

5207

-

5216

)

33

, , , , , .

ESMO Guidelines Working Group

Gastric marginal zone lymphoma of MALT type: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up.

,

Ann Oncol

,

2013

, vol.

24

suppl 6

(pg.

vi144

-

vi148

)

34

, , , et al.

Extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue of the salivary glands: a multicenter, international experience of 248 patients (IELSG 41).

,

Oncologist

,

2015

, vol.

20

10

(pg.

1149

-

1153

)

35

, , .

The risk of lymphoma development in autoimmune diseases: a meta-analysis.

,

Arch Intern Med

,

2005

, vol.

165

20

(pg.

2337

-

2344

)

36

, , , et al.

Medical history, lifestyle, family history, and occupational risk factors for marginal zone lymphoma: the InterLymph Non-Hodgkin Lymphoma Subtypes Project.

,

J Natl Cancer Inst Monogr

,

2014

, vol.

2014

48

(pg.

52

-

65

)

37

, .

Sjögren syndrome-associated lymphomas: an update on pathogenesis and management.

,

Br J Haematol

,

2015

, vol.

168

3

(pg.

317

-

327

)

38

, , , et al.

Associations of non-Hodgkin Lymphoma (NHL) risk with autoimmune conditions according to putative NHL loci.

,

Am J Epidemiol

,

2015

, vol.

181

6

(pg.

406

-

421

)

39

.

Apoptosis in autoimmune diseases.

,

Intern Med

,

2001

, vol.

40

4

(pg.

275

-

284

)

40

, , .

Mechanisms of autoantibody production and the relationship between autoantibodies and the clinical manifestations in Sjögren’s syndrome.

,

Transl Res

,

2006

, vol.

148

6

(pg.

281

-

288

)

41

, , , et al.

A genome-wide association study of marginal zone lymphoma shows association to the HLA region.

,

Nat Commun

,

2015

, vol.

6

pg.

5751

42

, , , , , .

Ongoing mutation in MALT lymphoma immunoglobulin gene suggests that antigen stimulation plays a role in the clonal expansion.

,

Leukemia

,

1996

, vol.

10

7

(pg.

1190

-

1197

)

43

, , , , .

Chronic inflammation and extra-nodal marginal-zone lymphomas of MALT-type.

,

Semin Cancer Biol

,

2014

, vol.

24

(pg.

33

-

42

)

44

, , , et al.

Gastric MALT lymphoma B cells express polyreactive, somatically mutated immunoglobulins.

,

Blood

,

2010

, vol.

115

3

(pg.

581

-

591

)

45

, , , et al.

Influence of antigen on the development of MALT lymphoma.

,

Blood

,

2006

, vol.

107

3

(pg.

1141

-

1148

)

46

, , , et al.

CD40 ligand and autoantigen are involved in the pathogenesis of low-grade B-cell lymphomas of mucosa-associated lymphoid tissue.

,

Dev Immunol

,

1998

, vol.

6

3-4

(pg.

187

-

195

)

47

, , , et al.

International Extranodal Lymphoma Study Group

Nongastric marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue.

,

Blood

,

2003

, vol.

101

7

(pg.

2489

-

2495

)

48

, , , et al.

Variable frequencies of MALT lymphoma-associated genetic aberrations in MALT lymphomas of different sites.

,

Leukemia

,

2004

, vol.

18

10

(pg.

1722

-

1726

)

49

, , , et al.

Mucosa-associated lymphoid tissue gastrointestinal and nongastrointestinal lymphoma behavior: analysis of 108 patients.

,

J Clin Oncol

,

1997

, vol.

15

4

(pg.

1624

-

1630

)

50

, , , et al.

Assessment of disease dissemination in gastric compared with extragastric mucosa-associated lymphoid tissue lymphoma using extensive staging: a single-center experience.

,

J Clin Oncol

,

2006

, vol.

24

19

(pg.

3136

-

3141

)

51

, , , et al.

Dissemination patterns in non-gastric MALT lymphoma.

,

Haematologica

,

2008

, vol.

93

2

(pg.

201

-

206

)

52

, , , et al.

Long-term outcome in localized extranodal mucosa-associated lymphoid tissue lymphomas treated with radiotherapy.

,

Cancer

,

2010

, vol.

116

16

(pg.

3815

-

3824

)

53

, , , .

Clinical outcomes and patterns of relapse in 320 patients with early and advanced-stage marginal zone lymphoma: the role of radiotherapy.

,

Hematol Oncol

,

2013

, vol.

31

S1

pg.

130

54

, , , et al.

MALT lymphoma in patients with autoimmune diseases: a comparative analysis of characteristics and clinical course.

,

Leukemia

,

2007

, vol.

21

8

(pg.

1812

-

1818

)

55

, , , et al.

Mucosa-associated lymphoid tissue lymphoma is a disseminated disease in one third of 158 patients analyzed.

,

Blood

,

2000

, vol.

95

3

(pg.

802

-

806

)

56

, ,

Aprile von Hohenstaufen

K

, et al.

Histologic transformation in marginal zone lymphomas.

,

Ann Oncol

,

2015

, vol.

26

11

(pg.

2329

-

2335

)

57

, , , et al.

Outcome of transformed marginal zone lymphomas treated in the rituximab era [abstract].

,

Blood

,

2015

, vol.

126

23

Abstract 5098

58

, , .

The management of nongastric MALT lymphomas.

,

Oncology (Williston Park)

,

2014

, vol.

28

1

(pg.

86

-

93

)

59

, , , et al.

ESMO Consensus conferences: guidelines on malignant lymphoma. part 2: marginal zone lymphoma, mantle cell lymphoma, peripheral T-cell lymphoma.

,

Ann Oncol

,

2013

, vol.

24

4

(pg.

857

-

877

)

60

, , , , , .

Detection rate of fluorine-18-fluorodeoxyglucose positron emission tomography in patients with marginal zone lymphoma of MALT type: a meta-analysis.

,

Hematol Oncol

,

2015

33(3):113-124

61

, , , et al.

Role of 18F-FDG-PET/CT in the management of marginal zone B cell lymphoma.

,

Hematol Oncol

,

2015

33(4):151-158

62

, , , et al.

Eradication of Borrelia burgdorferi infection in primary marginal zone B-cell lymphoma of the skin.

,

Hum Pathol

,

2000

, vol.

31

2

(pg.

263

-

268

)

63

, , , et al.

Evidence for an association between Chlamydia psittaci and ocular adnexal lymphomas.

,

J Natl Cancer Inst

,

2004

, vol.

96

8

(pg.

586

-

594

)

64

, , , et al.

Immunoproliferative small intestinal disease associated with Campylobacter jejuni.

,

N Engl J Med

,

2004

, vol.

350

3

(pg.

239

-

248

)

65

, , , et al.

Prevalence of Achromobacter xylosoxidans in pulmonary mucosa-associated lymphoid tissue lymphoma in different regions of Europe.

,

Br J Haematol

,

2014

, vol.

164

6

(pg.

804

-

810

)

66

, , , et al.

Prevalence of HCV infection in nongastric marginal zone B-cell lymphoma of MALT.

,

Ann Oncol

,

2007

, vol.

18

2

(pg.

346

-

350

)

67

, , , , , .

Indolent B-cell lymphomas associated with HCV infection: clinical and virological features and role of antiviral therapy.

,

Clin Dev Immunol

,

2012

, vol.

2012

638185

68

, .

Hepatitis C virus-associated B-cell non-Hodgkin lymphomas.

,

Hematology Am Soc Hematol Educ Program

,

2014

, vol.

2014

(pg.

590

-

598

)

69

, , , et al.

Effects of Helicobacter pylori eradication on early stage gastric mucosa-associated lymphoid tissue lymphoma.

,

Clin Gastroenterol Hepatol

,

2010

, vol.

8

2

(pg.

105

-

110

)

70

, .

Antibiotic therapy in nongastrointestinal MALT lymphoma: a review of the literature.

,

Blood

,

2013

, vol.

122

8

(pg.

1350

-

1357

)

71

, , , et al.

Six-month oral clarithromycin regimen is safe and active in extranodal marginal zone B-cell lymphomas: final results of a single-centre phase II trial.

,

Br J Haematol

,

2010

, vol.

150

2

(pg.

226

-

229

)

72

, , , et al.

High-dose clarithromycin is an active monotherapy for patients with relapsed/refractory extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue (MALT): the HD-K phase II trial.

,

Ann Oncol

,

2015

, vol.

26

8

(pg.

1760

-

1765

)

73

, , , .

Helicobacter pylori-associated gastritis and primary B-cell gastric lymphoma.

,

Lancet

,

1991

, vol.

338

8776

(pg.

1175

-

1176

)

74

, , , , , .

Helicobacter pylori and primary gastric lymphoma. A histopathologic and immunohistochemical analysis of 237 patients.

,

Cancer

,

1997

, vol.

79

1

(pg.

3

-

11

)

75

, , , et al.

Decreasing incidence of gastric MALT lymphomas in the era of anti-Helicobacter pylori interventions: results from a population-based study on extranodal marginal zone lymphomas.

,

Ann Oncol

,

2010

, vol.

21

4

(pg.

855

-

859

)

76

, , , et al.

Molecular analysis of the progression from Helicobacter pylori-associated chronic gastritis to mucosa-associated lymphoid-tissue lymphoma of the stomach.

,

N Engl J Med

,

1998

, vol.

338

12

(pg.

804

-

810

)

77

, .

Helicobacter pylori and mucosa-associated lymphoid tissue: what’s new.

,

Hematology Am Soc Hematol Educ Program

,

2013

, vol.

2013

(pg.

109

-

117

)

78

.

Helicobacter pylori CagA and gastric cancer: a paradigm for hit-and-run carcinogenesis.

,

Cell Host Microbe

,

2014

, vol.

15

3

(pg.

306

-

316

)

79

, , , , , .

MALT-type lymphoma of the stomach is associated with Helicobacter pylori strains expressing the CagA protein.

,

Gastroenterology

,

1997

, vol.

112

5

(pg.

1482

-

1486

)

80

, , , et al.

Gastric marginal zone lymphoma is associated with polymorphisms in genes involved in inflammatory response and antioxidative capacity.

,

Blood

,

2003

, vol.

102

3

(pg.

1007

-

1011

)

81

, , , et al.

A pooled analysis of three studies evaluating genetic variation in innate immunity genes and non-Hodgkin lymphoma risk.

,

Br J Haematol

,

2011

, vol.

152

6

(pg.

721

-

726

)

82

, , , et al.

Helicobacter pylori eradication as exclusive treatment for limited-stage gastric diffuse large B-cell lymphoma: results of a multicenter phase 2 trial.

,

Blood

,

2012

, vol.

120

18

(pg.

3858

-

3860

)

83

, , , 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

, vol.

119

21

(pg.

4838

-

4844

)

84

, , , et al.

Complete remission of primary high-grade B-cell gastric lymphoma after cure of Helicobacter pylori infection.

,

J Clin Oncol

,

2001

, vol.

19

7

(pg.

2041

-

2048

)

85

, , , et al.

Nuclear expression of BCL10 or nuclear factor kappa B predicts Helicobacter pylori-independent status of early-stage, high-grade gastric mucosa-associated lymphoid tissue lymphomas.

,

J Clin Oncol

,

2004

, vol.

22

17

(pg.

3491

-

3497

)

86

, , , et al.

Strong BCL10 nuclear expression identifies gastric MALT lymphomas that do not respond to H pylori eradication.

,

Gut

,

2006

, vol.

55

1

(pg.

137

-

138

)

87

, , , et al.

Long-term outcome following Helicobacter pylori eradication in a retrospective study of 105 patients with localized gastric marginal zone B-cell lymphoma of MALT type.

,

Ann Oncol

,

2009

, vol.

20

6

(pg.

1086

-

1093

)

88

, , , et al.

EGILS (European Gastro-Intestinal Lymphoma Study) Group

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

, vol.

56

12

(pg.

1685

-

1687

)

89

, , , et al.

JAPAN GAST Study Group

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

, vol.

61

4

(pg.

507

-

513

)

90

, , , et al.

Long-term follow-up of gastric MALT lymphoma after Helicobacter pylori eradication.

,

J Clin Oncol

,

2005

, vol.

23

31

(pg.

8018

-

8024

)

91

, , , et al.

Gastric MALT lymphoma: epidemiology and high adenocarcinoma risk in a nation-wide study.

,

Eur J Cancer

,

2008

, vol.

44

16

(pg.

2470

-

2476

)

92

, , , et al.

Second cancers and residual disease in patients treated for gastric mucosa-associated lymphoid tissue lymphoma by Helicobacter pylori eradication and followed for 10 years.

,

Gastroenterology

,

2012

143(4):936-942

93

, , , et al.

Eradication therapy in Helicobacter pylori-negative, gastric low-grade mucosa-associated lymphoid tissue lymphoma patients: a systematic review.

,

J Clin Gastroenterol

,

2013

, vol.

47

10

(pg.

824

-

827

)

94

, , , et al.

Chlamydophila psittaci eradication with doxycycline as first-line targeted therapy for ocular adnexae lymphoma: final results of an international phase II trial.

,

J Clin Oncol

,

2012

, vol.

30

24

(pg.

2988

-

2994

)

95

, , , et al.

Chlamydia infection and lymphomas: association beyond ocular adnexal lymphomas highlighted by multiple detection methods.

,

Clin Cancer Res

,

2008

, vol.

14

18

(pg.

5794

-

5800

)

96

, , , et al.

A woman and her canary: a tale of chlamydiae and lymphomas.

,

J Natl Cancer Inst

,

2007

, vol.

99

18

(pg.

1418

-

1419

)

97

, , , et al.

Bacteria-eradicating therapy with doxycycline in ocular adnexal MALT lymphoma: a multicenter prospective trial.

,

J Natl Cancer Inst

,

2006

, vol.

98

19

(pg.

1375

-

1382

)

98

, , , et al.

First-line therapy with doxycycline in ocular adnexal mucosa-associated lymphoid tissue lymphoma: a retrospective analysis of clinical predictors.

,

Cancer Sci

,

2010

, vol.

101

5

(pg.

1199

-

1203

)

99

, , .

Marginal zone lymphomas and infectious agents.

,

Semin Cancer Biol

,

2013

, vol.

23

6

(pg.

431

-

440

)

100

, , , , , . , , , et al.

Extranodal marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue (MALT lymphoma).

,

WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues

Lyon, France: IARC; 2008:214-217

101

, .

Immunoproliferative small intestinal disease (IPSID): a model for mature B-cell neoplasms.

,

Blood

,

2005

, vol.

105

6

(pg.

2274

-

2280

)

102

, , , et al.

A retrospective international study on primary extranodal marginal zone lymphoma of the lung (BALT lymphoma) on behalf of International Extranodal Lymphoma Study Group (IELSG) [published online ahead of print July 7, 2015].

,

Hematol Oncol

103

, , , et al.

Chlamydiae and Mycoplasma infections in pulmonary MALT lymphoma.

,

Br J Cancer

,

2007

, vol.

97

7

(pg.

949

-

951

)

104

, , , et al.

Extranodal marginal zone lymphoma of the lung: evolution from an underlying reactive lymphoproliferative disorder.

,

J Clin Exp Pathol

,

2015

, vol.

5

1

pg.

208

105

, , , et al.

ANRS HC-13 Lympho-C Study Group

Antiviral therapy is associated with a better survival in patients with hepatitis C virus and B-cell non-Hodgkin lymphomas, ANRS HC-13 lympho-C study.

,

Am J Hematol

,

2015

, vol.

90

3

(pg.

197

-

203

)

106

, , , et al.

Anti-lymphoma activity of interferon-free antiviral treatment in patients with indolent B-cell lymphomas associated with hepatitis C virus infection [abstract].

,

Blood

,

2015

, vol.

126

23

Abstract 3938

107

, .

Hepatitis viruses and non-Hodgkin lymphoma: epidemiology, mechanisms of tumorigenesis, and therapeutic opportunities.

,

Blood

,

2011

, vol.

117

6

(pg.

1792

-

1798

)

108

, , , et al.

Most cases of primary salivary mucosa-associated lymphoid tissue lymphoma are associated either with Sjoegren syndrome or hepatitis C virus infection.

,

Br J Haematol

,

2004

, vol.

126

1

(pg.

43

-

49

)

109

, , .

Disappearance of gastric mucosa-associated lymphoid tissue in hepatitis C virus-positive patients after anti-hepatitis C virus therapy.

,

J Clin Gastroenterol

,

2004

, vol.

38

4

(pg.

360

-

363

)

110

, , , .

Lacrimal gland marginal zone lymphoma: regression after treatment of chronic hepatitis C virus infection: case report and review of the literature.

,

Intern Med

,

2013

, vol.

52

23

(pg.

2615

-

2618

)

111

, , , et al.

Subcutaneous ‘lipoma-like’ B-cell lymphoma associated with HCV infection: a new presentation of primary extranodal marginal zone B-cell lymphoma of MALT.

,

Ann Oncol

,

2010

, vol.

21

6

(pg.

1189

-

1195

)

112

, , , et al.

Outcome in relation to treatment modalities in 48 patients with localized gastric MALT lymphoma: a retrospective study of patients treated during 1976-2001.

,

Leuk Lymphoma

,

2003

, vol.

44

2

(pg.

257

-

262

)

113

, , , et al.

Clinical features, treatment and outcome in a series of 93 patients with low-grade gastric MALT lymphoma.

,

Leuk Lymphoma

,

1997

, vol.

26

5-6

(pg.

527

-

537

)

114

, , , et al.

Retrospective comparison of the effectiveness of various treatment modalities of extragastric MALT lymphoma: a single-center analysis.

,

Ann Hematol

,

2014

, vol.

93

8

(pg.

1287

-

1295

)

115

, , , et al.

Long-term outcome for gastric marginal zone lymphoma treated with radiotherapy: a retrospective, multi-centre, International Extranodal Lymphoma Study Group study.

,

Ann Oncol

,

2013

, vol.

24

5

(pg.

1344

-

1351

)

116

, .

Radiation therapy for localized low-grade non-Hodgkin’s lymphomas.

,

Hematol Oncol

,

2005

, vol.

23

1

(pg.

10

-

17

)

117

, , , et al.

Treatment results in localized primary gastric lymphoma: data of patients registered within the German multicenter study (GIT NHL 02/96).

,

J Clin Oncol

,

2005

, vol.

23

28

(pg.

7050

-

7059

)

118

National Comprehensive Cancer Network (NCCN)

National Comprehensive Cancer Network guidelines V.2.2013: non-Hodgkin’s lymphoma.

119

, , , et al.

MALT lymphomas: pathogenesis can drive treatment.

,

Oncology (Williston Park)

,

2011

, vol.

25

12

(pg.

1134

-

1142, 1147

)

120

, , , et al.

Clinical activity of rituximab in extranodal marginal zone B-cell lymphoma of MALT type.

,

Blood

,

2003

, vol.

102

8

(pg.

2741

-

2745

)

121

, , , et al.

Clinical activity of rituximab in gastric marginal zone non-Hodgkin’s lymphoma resistant to or not eligible for anti-Helicobacter pylori therapy.

,

J Clin Oncol

,

2005

, vol.

23

9

(pg.

1979

-

1983

)

122

, , , et al.

Chlorambucil plus rituximab produces better event-free and progression-free survival in comparison with chlorambucil or rituximab alone in extranodal marginal zone B-cell lymphoma (MALT lymphoma): results of the IELSG-19 study.

,

Hematol Oncol

,

2013

, vol.

31

S1

(pg.

97

-

98

)

123

, , , et al.

Addition of rituximab to chlorambucil produces superior event-free survival in the treatment of patients with extranodal marginal-zone B-cell lymphoma: 5-year analysis of the IELSG-19 Randomized Study.

,

J Clin Oncol

,

2013

, vol.

31

5

(pg.

565

-

572

)

124

, , , et al.

Bendamustine plus rituximab in first line systemic treatment for extranodal MALT lymphoma: final results of phase II trial of the Spanish lymphoma study group (GELTAMO).

,

Hematol Oncol

,

2013

, vol.

31

S1

(pg.

129

-

130

)

125

, , , et al.

A phase 2 study of concurrent fludarabine and rituximab for the treatment of marginal zone lymphomas.

,

Br J Haematol

,

2009

, vol.

145

6

(pg.

741

-

748

)

126

, , , et al.

Safety and efficacy of abexinostat, a pan-histone deacetylase (HDAC) inhibitor, in non-Hodgkin lymphoma and chronic lymphocytic leukemia: results of an ongoing phase 2 study [abstract].

,

Blood

,

2015

, vol.

126

23

Abstract 256

127

, , , et al.

International Extranodal Lymphoma Study Group (IELSG)

Clinical activity of bortezomib in relapsed/refractory MALT lymphomas: results of a phase II study of the International Extranodal Lymphoma Study Group (IELSG).

,

Ann Oncol

,

2011

, vol.

22

3

(pg.

689

-

695

)

128

, , , et al.

Phase 2 trial of entospletinib (GS-9973), a selective Syk inhibitor, in indolent non-Hodgkin’s lymphoma (iNHL) [abstract].

,

Blood

,

2015

, vol.

126

23

Abstract 1545

129

, , , et al.

Clinical activity of everolimus in relapsed/refractory marginal zone B-cell lymphomas: results of a phase II study of the International Extranodal Lymphoma Study Group.

,

Br J Haematol

,

2014

, vol.

166

1

(pg.

69

-

76

)

130

, , , et al.

Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignancies.

,

J Clin Oncol

,

2013

, vol.

31

1

(pg.

88

-

94

)

131

, , , et al.

PI3Kδ inhibition by idelalisib in patients with relapsed indolent lymphoma.

,

N Engl J Med

,

2014

, vol.

370

11

(pg.

1008

-

1018

)

132

, , , et al.

Idelalisib monotherapy and durable responses in patients with relapsed or refractory marginal zone lymphoma (MZL) [abstract].

,

Blood

,

2015

, vol.

126

23

Abstract 1543

133

, , , et al.

A phase II study of lenalidomide in patients with extranodal marginal zone B-cell lymphoma of the mucosa associated lymphoid tissue (MALT lymphoma).

,

Haematologica

,

2013

, vol.

98

3

(pg.

353

-

356

)

134

, , , et al.

Phase 1 study of tazemetostat (EPZ-6438), an inhibitor of enhancer of zeste-homolog 2 (EZH2): preliminary safety and activity in relapsed or refractory non-Hodgkin lymphoma (NHL) patients [abstract].

,

Blood

,

2015

, vol.

126

23

Abstract 473

135

, , .

Absence of efficacy of thalidomide monotherapy in patients with extranodal marginal zone B-cell lymphoma of the mucosa-associated lymphoid tissue (MALT lymphoma).

,

Ann Oncol

,

2009

, vol.

20

8

(pg.

1446

-

1447

)

136

, , , et al.

The combination of weekly infusion of rituximab and bortezomib is effective in relapsed or refractory indolent and mantle cell lymphoma: long-term results of phase II BRIL06 study of the Fondazione Italiana Linfomi (FIL) [abstract].

,

Blood

,

2015

, vol.

126

23

Abstract 2735

137

, , , et al.

Updated results of a phase I study of ibrutinib and lenalidomide in patients with relapsed and refractory B-cell non-Hodgkin’s lymphoma [abstract].

,

Blood

,

2015

, vol.

126

23

Abstract 3983

138

, , , et al.

A phase 1/1b study of rituximab, bendamustine, and ibrutinib in patients with untreated and relapsed/refractory non-Hodgkin lymphoma.

,

Blood

,

2015

, vol.

125

2

(pg.

242

-

248

)

139

, , , , , .

Lenalidomide, idelalisib, and rituximab are unacceptably toxic in patients with relapsed/refractory indolent lymphoma.

,

Blood

,

2015

, vol.

125

21

(pg.

3357

-

3359

)

140

, , , , , .

AGMT MALT-2: a phase II study of rituximab plus lenalidomide in patients with extranodal marginal zone B-cell lymphoma of the mucosa-associated lymphoid tissue (MALT lymphoma) [abstract].

,

Blood

,

2015

, vol.

126

23

Abstract 3973

141

, , , et al.

Safety and activity of lenalidomide and rituximab in untreated indolent lymphoma: an open-label, phase 2 trial.

,

Lancet Oncol

,

2014

, vol.

15

12

(pg.

1311

-

1318

)

142

, .

A phase I study of bendamustine, lenalidomide and rituximab in relapsed and refractory lymphomas.

,

Br J Haematol

,

2015

, vol.

169

4

(pg.

528

-

533

)

143

, , , et al.

A dose-escalation study of venetoclax (ABT-199/GDC-0199) in combination with bendamustine and rituximab in patients with relapsed or refractory non-Hodgkin’s lymphoma [abstract].

,

Blood

,

2015

, vol.

126

23

Abstract 255

144

, , , , , .

Delayed efficacy after treatment with lenalidomide or thalidomide in patients with mucosa-associated lymphoid tissue lymphoma.

,

Oncologist

,

2016

21(1):72-75

145

.

Clinical presentation and management of marginal zone lymphomas.

,

Hematology Am Soc Hematol Educ Program

,

2005

, vol.

2005

(pg.

307

-

313

)

146

, , , et al.

ESMO Guidelines Committee

Diffuse large B-cell lymphoma (DLBCL): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up.

,

Ann Oncol

,

2015

, vol.

26

suppl 5

(pg.

v116

-

v125

)

147

, , .

Recent developments in nongastric mucosa-associated lymphoid tissue lymphoma.

,

Curr Hematol Malig Rep

,

2011

, vol.

6

4

(pg.

216

-

221

)

Author notes

*

E.Z. and F.B. equally contributed to this manuscript.

© 2016 by The American Society of Hematology

2016

Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.