Non-Hodgkin lymphomas develop from nodal and extranodal lymphoid tissues. A distinct subset of extranodal lymphomas arising from B cells of the marginal zone (MZ) of mucosa-associated lymphoid tissue (MALT) or spleen has been individualized. Growing evidence indicates that MZ lymphomas are associated with chronic antigenic stimulation by microbial pathogens and/or autoantigens. The list of microbial species associated with MZ lymphoproliferations has grown longer with molecular investigations and now comprises at least 5 distinct members: H pylori, C jejuni, B burgdorferi, C psittaci, and hepatitis C virus (HCV), which have been associated with gastric lymphoma, immunoproliferative small intestinal disease, cutaneous lymphoma, ocular lymphoma, and spleen lymphoma, respectively. A pathophysiologic scenario involving chronic and sustained stimulation of the immune system leading to lymphoid transformation has emerged. It defines a distinct category of infection-associated lymphoid malignancies, in which the infectious agent does not directly infect and transform lymphoid cells, as do the lymphotropic oncogenic viruses Epstein-Barr virus (EBV), human herpesvirus 8 (HHV8), and human T-lymphotropic virus 1 (HTLV-1), but rather indirectly increases the probability of lymphoid transformation by chronically stimulating the immune system to maintain a protracted proliferative state.

The geographic heterogeneity in the incidence of B-cell non-Hodgkin lymphomas (NHLs) suggests that environmental factors such as infections might have a role in lymphomagenesis.1,2 

Because of the inherent genetic instability of lymphocytes, lymphoid proliferation increases the risk of transformation, and sustained activation of the lymphoid system, which can be observed during chronic infection, immunodeficiency, and autoimmunity, constitutes a risk factor for lymphomas.3-5  Congenital and acquired immunodeficiencies associated with HIV infection and solid organ or hematopoietic transplantation increase the risk of developing B-cell NHLs.6,7  Similarly, Sjögren syndrome and other autoimmune conditions are also associated with an increased risk of lymphomas.8,9 

Certain types of lymphomas are associated with specific microbial infections, and infection-associated lymphomas currently fall in diverse histopathologic categories (Table 1).10  Infections may contribute to lymphomagenesis by promoting favorable conditions for lymphocyte transformation, such a increased proliferation or decreased apoptosis of lymphoid cells.11 

Table 1.

World Health Organization classification lymphoma subtypes associated with infections


Microbial pathogen

World Health Organization (WHO) histologic subtype
Human T-lymphotropic virus 1 (HTLV-1)   Adult T-cell leukemia/lymphoma  
Human immunodeficiency virus (HIV)   Hodgkin disease (with EBV)  
  Burkitt lymphoma (with or without EBV)  
  DLBCL (including primary effusion lymphoma and plasmablastic lymphoma)  
  Extranodal MZ lymphoma, MALT-type (rare)  
  T-cell lymphoma (rare)  
Epstein-Barr virus (EBV)   Hodgkin disease  
  Polymorphic PTLD  
  Burkitt lymphoma  
  Monomorphic PTLD (DLBCL)  
  Primary effusion lymphoma (with HHV8)  
Human herpesvirus 8/Kaposi sarcoma-associated herpesvirus (HHV8/KSHV)   Primary effusion lymphoma  
  Plasmablastic lymphoma (DLBCL)  
  PTLD (rare)  
Hepatitis C virus (HCV)   SLVL (splenic MZ lymphoma)  
  Other marginal zone lymphoma  
  DLBCL  
Helicobacter pylori  Gastric MALT lymphoma (extranodal MZ lymphoma, MALT-type)  
Campylobacter jejuni  IPSID (extranodal MZ lymphoma, MALT-type)  
Borrelia burgdorferi  Primary cutaneous B-cell lymphoma (various WHO subtypes including extranodal MZ lymphoma, MALT-type)  
Chlamydia psittaci
 
Ocular adnexal lymphoma (extranodal MZ lymphoma, MALT-type)
 

Microbial pathogen

World Health Organization (WHO) histologic subtype
Human T-lymphotropic virus 1 (HTLV-1)   Adult T-cell leukemia/lymphoma  
Human immunodeficiency virus (HIV)   Hodgkin disease (with EBV)  
  Burkitt lymphoma (with or without EBV)  
  DLBCL (including primary effusion lymphoma and plasmablastic lymphoma)  
  Extranodal MZ lymphoma, MALT-type (rare)  
  T-cell lymphoma (rare)  
Epstein-Barr virus (EBV)   Hodgkin disease  
  Polymorphic PTLD  
  Burkitt lymphoma  
  Monomorphic PTLD (DLBCL)  
  Primary effusion lymphoma (with HHV8)  
Human herpesvirus 8/Kaposi sarcoma-associated herpesvirus (HHV8/KSHV)   Primary effusion lymphoma  
  Plasmablastic lymphoma (DLBCL)  
  PTLD (rare)  
Hepatitis C virus (HCV)   SLVL (splenic MZ lymphoma)  
  Other marginal zone lymphoma  
  DLBCL  
Helicobacter pylori  Gastric MALT lymphoma (extranodal MZ lymphoma, MALT-type)  
Campylobacter jejuni  IPSID (extranodal MZ lymphoma, MALT-type)  
Borrelia burgdorferi  Primary cutaneous B-cell lymphoma (various WHO subtypes including extranodal MZ lymphoma, MALT-type)  
Chlamydia psittaci
 
Ocular adnexal lymphoma (extranodal MZ lymphoma, MALT-type)
 

DLBCL indicates diffuse large B-cell lymphoma; MZ, marginal zone; MALT, mucosa-associated lymphoid tissue; PTLD, posttransplantation lymphoproliferative disorder; SLVL, splenic lymphoma with villous lymphocytes; and IPSID, immunoproliferative small intestinal disease.

Direct lymphocyte transformation by a given microbial agent is the simplest scenario accounting for infection-associated lymphomas. Lymphotropic transforming viruses such as Epstein-Barr virus (EBV), human herpesvirus 8 (HHV8), and human T-lymphotropic virus 1 (HTLV-1) directly infect a subset of lymphoid cells in which they express viral oncogenes.12-14 

An alternative scenario to direct transformation of lymphocytes has more recently emerged for microbial species associated with lymphomas but that do not directly infect or transform lymphoid cells. They have in common the ability to persist chronically in host tissues and trigger a sustained lymphoid proliferation, giving a selective advantage to lymphoid clones that still remain dependent upon antigen stimulation.15,16  According to this model, the microbial pathogen is neither intrinsically transforming nor oncogenic, but can be viewed as a chronic source of antigens increasing the proliferative rate of lymphoid effectors, hence fueling the transformation process.

This model has emerged with the description of several lymphomas developing in the context of chronic antigen-dependent immune stimulation, among which H pylori–associated gastric mucosa-associated lymphoid tissue (MALT) lymphoma is the best characterized15,17,18  (Figure 1). Precise elucidation of the mechanisms underlying this “indirect” lymphomagenesis as well as completion of the inventory of the microbial species driving these antigen-dependent lymphoproliferations may provide important clues for their early diagnosis and the rationalization of the therapeutic interventions for this subtype of lymphomas.

This group of lymphomas often involves extranodal sites—normally devoid of organized lymphoid tissue—and manifests initially as indolent low-grade proliferations, reminiscent of the normal lymphoid hyperplasia driven by a physiologic antigenic stimulation.19-21  With the progression of the disease, additional oncogenic events may occur—such as constitutive activation of signaling pathways following chromosomal translocations or inactivation of tumor suppressor genes by hypermethylation or mutations—leading the lymphoproliferation to become independent of antigenic stimulation.22 

Figure 1.

H pylori and gastric MALT lymphomagenesis paradigm for infection-associated indirect lymphoid transformation. (A) Persisting antigens (Ag's) elicit a polyclonal B-cell response. Costimulation is provided by cytokines and members of the tumor necrosis factor superfamily (CD40-CD40L in T-cell–dependent responses and B-cell activating factor [BAFF]/B-lymphocyte stimulator [BLyS] or a proliferation-inducing ligand [APRIL] produced by dendritic cells in T-cell–independent responses). In the case of H pylori, T cells specific for H pylori epitopes provide help to B cells that recognize cross-reactive autoantigens present in the gastric mucosa such as fucosylated sialyl–Lewis X through CD40-CD40L costimulation. (B) Occurrence of genetic events such as p15 and p16 hypermethylation provide a selective advantage leading to the outgrowth of an antigen-responsive clone. Antigen dependence reflects the requirement for BCR signals such as NF-κB activation (see also Figure 5). (C) Progression toward antigen-independent (therefore antimicrobial-insensitive) MALT lymphoma is associated with the occurrence of additional genetic events. Chromosomal translocations involving MALT1 and Bcl-10 lead to a constitutive activation of NF-κB, bypassing the requirement for BCR-dependent signals (Figure 5). The t(11;18) translocation occurs early during B-cell proliferation and accelerates the transformation process in an antigen-independent fashion. The t(1;14) and t(14;18) translocations also lead to BCR-dependent NF-κB activation, but occur later after an antigen-dependent phase and can be associated with additional cytogenetic abnormalities. The t(3;14) translocation produces an IgH-FoxP1 transcript in 10% of MALT lymphomas that do not harbor other translocations involving the MALT1/Bcl-10 pathway. The precise role of t(3;14) is not known. P53 mutation is associated with transformation to high-grade lymphoma.

Figure 1.

H pylori and gastric MALT lymphomagenesis paradigm for infection-associated indirect lymphoid transformation. (A) Persisting antigens (Ag's) elicit a polyclonal B-cell response. Costimulation is provided by cytokines and members of the tumor necrosis factor superfamily (CD40-CD40L in T-cell–dependent responses and B-cell activating factor [BAFF]/B-lymphocyte stimulator [BLyS] or a proliferation-inducing ligand [APRIL] produced by dendritic cells in T-cell–independent responses). In the case of H pylori, T cells specific for H pylori epitopes provide help to B cells that recognize cross-reactive autoantigens present in the gastric mucosa such as fucosylated sialyl–Lewis X through CD40-CD40L costimulation. (B) Occurrence of genetic events such as p15 and p16 hypermethylation provide a selective advantage leading to the outgrowth of an antigen-responsive clone. Antigen dependence reflects the requirement for BCR signals such as NF-κB activation (see also Figure 5). (C) Progression toward antigen-independent (therefore antimicrobial-insensitive) MALT lymphoma is associated with the occurrence of additional genetic events. Chromosomal translocations involving MALT1 and Bcl-10 lead to a constitutive activation of NF-κB, bypassing the requirement for BCR-dependent signals (Figure 5). The t(11;18) translocation occurs early during B-cell proliferation and accelerates the transformation process in an antigen-independent fashion. The t(1;14) and t(14;18) translocations also lead to BCR-dependent NF-κB activation, but occur later after an antigen-dependent phase and can be associated with additional cytogenetic abnormalities. The t(3;14) translocation produces an IgH-FoxP1 transcript in 10% of MALT lymphomas that do not harbor other translocations involving the MALT1/Bcl-10 pathway. The precise role of t(3;14) is not known. P53 mutation is associated with transformation to high-grade lymphoma.

Close modal

Although these lymphomas are associated with diverse microbial species, they all appear to originate from “marginal zone” (MZ) lymphocytes. These cells are anatomically positioned in the lymphoid organs (spleen and lymph nodes) and in the MALT to function as a first line of defense against invading pathogens.23,24  Furthermore, the low activation threshold of these cells may predispose them to neoplastic transformation.16,25 

Mature B cells are heterogeneous with respect to their microanatomic location in the lymphoid organs and functional properties (Figure 2). Follicular (FO) B cells (IgMlowIgDhighCD21interCD23high) constitute the major subset of B cells and participate in T-cell–dependent (TD) immune responses in the germinal center (GC), where they receive help from antigen-specific T cells (ie, cognate interactions) through CD40-CD40L engagement. The GC reaction leads to isotype class-switched memory B cells (IgMIgDCD27+) with somatic hypermutations (SHMs) in their immunoglobulin gene segments. Upon antigenic rechallenge, they rapidly undergo terminal differentiation into plasma cells producing large amounts of high-affinity antibodies.

Figure 2.

Organization of the lymphoid organs and B-cell responses. Top: Schematic view of a Peyer patch (left) and lymphoid follicles in the spleen (right) surrounded by the arteriolar marginal sinus. MZ indicates marginal zone; GC, germinal center; DC, dendritic cell; and FDC, follicular dendritic cell. The marginal zone is less conspicuous in the lymph nodes. Bottom left: Antigens having access to B-cell follicles are presented to B cells by FDCs. Antigen-specific B cells are activated by their BCR (signal 1) and proliferate, leading to the formation of the GC. Costimulation (signal 2) is provided by T-cell–derived cytokines and CD40L-CD40 interactions. GC T cells also express FasL, and autoreactive B cells (expressing Fas) are deleted by FasL-Fas interactions. Bottom right: Antigens captured in the blood by dendritic cells or directly accessing the MZ are presented to MZ B cells. MZ B cells are activated by BCR signal (signal 1). Costimulation (signal 2) is provided by DC-derived cytokines and the tumor necrosis factor superfamily member BAFF/BLyS or APRIL. Activated MZ B cells rapidly differentiate into plasma cells that secrete large amounts of IgM. In MZ B cells, mutations in immunoglobulin genes are acquired either during ontogeny in an antigen-independent fashion26  or during T-cell–dependent responses occurring in the GC.27 

Figure 2.

Organization of the lymphoid organs and B-cell responses. Top: Schematic view of a Peyer patch (left) and lymphoid follicles in the spleen (right) surrounded by the arteriolar marginal sinus. MZ indicates marginal zone; GC, germinal center; DC, dendritic cell; and FDC, follicular dendritic cell. The marginal zone is less conspicuous in the lymph nodes. Bottom left: Antigens having access to B-cell follicles are presented to B cells by FDCs. Antigen-specific B cells are activated by their BCR (signal 1) and proliferate, leading to the formation of the GC. Costimulation (signal 2) is provided by T-cell–derived cytokines and CD40L-CD40 interactions. GC T cells also express FasL, and autoreactive B cells (expressing Fas) are deleted by FasL-Fas interactions. Bottom right: Antigens captured in the blood by dendritic cells or directly accessing the MZ are presented to MZ B cells. MZ B cells are activated by BCR signal (signal 1). Costimulation (signal 2) is provided by DC-derived cytokines and the tumor necrosis factor superfamily member BAFF/BLyS or APRIL. Activated MZ B cells rapidly differentiate into plasma cells that secrete large amounts of IgM. In MZ B cells, mutations in immunoglobulin genes are acquired either during ontogeny in an antigen-independent fashion26  or during T-cell–dependent responses occurring in the GC.27 

Close modal

The marginal zone (MZ), which surrounds B-cell follicles in the spleen and in extranodal lymphoid tissue, contains a distinct subset of B cells, the MZ B cells (IgMhighIgDlowCD21highCD23low/–).28  MZ B cells participate in T-cell–independent (TI) “innatelike” immune responses to microbial pathogens,29  and can rapidly proliferate and differentiate into IgM or even switch to other isotype-secreting plasma cells, producing the bulk of the primary antibody response.30  TI responses do not generate memory B cells, consistent with a relatively short-lived antibody production. MZ B cells can be viewed as a bridge between the innate and adaptive immune responses to pathogens invading the host. In contrast to rodents, most human MZ B cells are somatically mutated (IgM+IgD+CD27+).26  It is still debated if these mutated MZ B cells in humans are in fact memory B cells originating from TD responses occurring in the GC and later homing to the MZ,27,31  or whether they are generated through a preimmune, antigen-independent diversification pathway.26  There is evidence that these mutated MZ B cells could be positively selected by autoantigens during B-cell ontogeny.32  Autoantigens or commensal bacterial antigens could be candidates for such selection processes occurring outside of the GC, and possibly involving CD40-CD40L cognate help by natural killer T (NKT) cells30  or BAFF-mediated noncognate help by MZ-resident macrophages or dendritic cells.28,33  Both B-cell subsets cooperate during the immune response to microbial polysaccharides: MZ B cells constitute the initial rapid response and can efficiently prime CD4 T cells that will subsequently provide costimulation to GC B cells.28  MZ B cells can also participate, though less efficiently than FO B cells, in TD antibody responses.28  Not surprisingly, owing to their frequent autoreactive and cross-reactive repertoire and to their relative hyperreactivity to activation, these cells are found in various pathologic conditions involving autoimmunity and infection. Lymphomas arising from MZ B cells can thus be expected to retain some of their seminal features, as explained in “Indirect transformation of lymphoid cells by microbial pathogens.”

EBV is associated with a number of B-cell malignancies, including Burkitt lymphoma (BL), Hodgkin disease (HD), posttransplantation lymphoproliferative disorder (PTLD), as well as a subset of T- and NK-cell malignant proliferations,12  and frequently arises in the course of an underlying immunodeficiency. EBV infects, immortalizes, and transforms B cells in vitro and establishes a persistent latent infection34,35  (Figure 3). Viral genes expressed during latency subvert normal proliferation and survival pathways.36  One of the major oncogenes, the latent membrane protein 1 (LMP1), functions as a constitutively active member of the TNF-R family, closely related to CD40, a receptor whose engagement on normal B cells leads to B-cell activation.37  LMP1 expression is required for proliferation and transformation and is critical for in vitro immortalization of B cells.38,39  LMP2A is another EBV latent protein that can mimic survival signals from the B-cell receptor and rescue B cells lacking surface immunoglobulin.40,41  In HD, the malignant Reed-Sternberg cells are thought to derive from GC B cells that have undergone extensive crippling mutations of their immunoglobulin genes, precluding the expression of a functional surface receptor. Normal GC B cells, in this situation, would be eliminated by apoptosis, and one possible role for EBV in the pathogenesis of HD would be to provide proliferative and antiapoptotic signals (through LMP1 and LMP2) to “crippled” GC B cells, enabling them to escape apoptosis.42 

Figure 3.

EBV infection and direct transformation of B cells. EBV infects naive B cells through 2 surface receptors, CD21 and the class II MHC, HLA-DQ. During latency, the virus is maintained as an episome in the nucleus of infected cells, and viral genes are expressed, in the absence of lytic replication of the virus. EBV subverts normal B-cell differentiation, notably by the expression of LMP1, a viral latent protein expressed at the surface of infected cells. LMP1 associates with transduction molecules such as TRADD and TRAF and activates the NF-κB pathway in a CD40-like manner. LMP1 is required for the activation and immortalization of B cells. LMP2 is a viral transmembrane protein that associates with Lyn/Syk kinase and PI3 kinase, leading to the activation of PKC and AKT, respectively. LMP2 can substitute for signals emanating from the BCR. Both LMP1 and LMP2 converge to activate proliferation and survival pathways in EBV latently infected cells. EBNA2 transactivates LMP1 and a number of cellular genes involved in activation and proliferation. Polyclonal infected B cells proliferate and produce immortalized lymphoblastoid cell lines in vitro. In vivo, EBV-infected B cells are negatively controlled by anti-EBV cytotoxic T lymphocytes (CTLs). Failure to control EBV-infected B cells may lead to the development of posttransplantation lymphoproliferative disorder (PTLD). Additional oncogenic mutations lead to clonal selection and evolution toward monoclonal tumors such as Burkitt lymphoma (BL), Hodgkin disease (HD), and diffuse large B-cell lymphomas (DLBCLs) in immunocompromised patients.

Figure 3.

EBV infection and direct transformation of B cells. EBV infects naive B cells through 2 surface receptors, CD21 and the class II MHC, HLA-DQ. During latency, the virus is maintained as an episome in the nucleus of infected cells, and viral genes are expressed, in the absence of lytic replication of the virus. EBV subverts normal B-cell differentiation, notably by the expression of LMP1, a viral latent protein expressed at the surface of infected cells. LMP1 associates with transduction molecules such as TRADD and TRAF and activates the NF-κB pathway in a CD40-like manner. LMP1 is required for the activation and immortalization of B cells. LMP2 is a viral transmembrane protein that associates with Lyn/Syk kinase and PI3 kinase, leading to the activation of PKC and AKT, respectively. LMP2 can substitute for signals emanating from the BCR. Both LMP1 and LMP2 converge to activate proliferation and survival pathways in EBV latently infected cells. EBNA2 transactivates LMP1 and a number of cellular genes involved in activation and proliferation. Polyclonal infected B cells proliferate and produce immortalized lymphoblastoid cell lines in vitro. In vivo, EBV-infected B cells are negatively controlled by anti-EBV cytotoxic T lymphocytes (CTLs). Failure to control EBV-infected B cells may lead to the development of posttransplantation lymphoproliferative disorder (PTLD). Additional oncogenic mutations lead to clonal selection and evolution toward monoclonal tumors such as Burkitt lymphoma (BL), Hodgkin disease (HD), and diffuse large B-cell lymphomas (DLBCLs) in immunocompromised patients.

Close modal

HHV8 is closely related to EBV and is also associated with a number of B-cell lymphoproliferative disorders including multicentric Castleman disease and primary effusion and plasmablastic lymphomas.43  As does EBV, HHV8 encodes several genes that interfere with cell-signaling pathways involved in proliferation and survival, and that may play a role in cellular transformation (for review see Damania41 ).

The human oncogenic retrovirus HTLV-1 infects and immortalizes CD4+ T cells. The Tax oncoprotein interferes with numerous cell-signaling pathways and is thought to play a major role in immortalization.14,44 

In general, the relationship between viral cycle and viral oncogenesis is complex. As a retrovirus, HTLV-1 integrates in the genome of infected cells. Although HTLV-1 replication is present in infected individuals, leukemic cells in adult T-cell leukemia (ATL) harbor latent integrated virus. The genomes of EBV and HHV8 are maintained as episomes in latently infected cells, and viral replication is not required for B-cell transformation of EBV-infected cells. Although HHV8 also establishes a latent infection in B cells, there is evidence that lytic replication is implicated in the early steps of oncogenesis.43  Since antiviral drugs target replication, they are mostly ineffective against virus-associated lymphoproliferations. HTLV-1, however, stands out as an exception because antiretroviral therapy with azidothymidine and high-dose interferon alfa has shown to be effective, at least in previously untreated ATL,14  although these drugs may also act at different levels by modulating cellular and viral gene expression such as inhibition of NF-κB and up-regulation of viral genes.45 

Since the identification of the role of Helicobacter pylori in the pathogenesis of gastric MALT lymphoma, several other low-grade B-cell lymphomas have been associated with chronic infections. Strikingly, most if not all of these lymphomas are derived from MZ B cells.

The International Lymphoma Study Group now recognizes 3 distinct lymphoma entities deriving from MZ B lymphocytes10,46 : (1) splenic MZ lymphomas, (2) extranodal MZ lymphoma of MALT-type, and (3) nodal MZ lymphomas. Although many aspects of their histology and molecular pathogenesis are distinct, these entities share a number of common features, the most striking being their possible association with chronic antigenic stimulation by microbial and/or autoantigens.

MALT lymphomas

Low-grade lymphomas originating from the MALT were originally described in the stomach and small intestine.47,48  They may also develop in other mucosal sites such as the salivary and lacrimal glands, the thyroid, and the bronchi. They have now all been classified in the same nosologic entity generically called “MALT lymphoma.”15,49,50  The majority of the organs in which MALT lymphomas develop are normally devoid of lymphoid tissue, and, in most cases, MALT acquisition is induced prior to the development of lymphoma, as a response to a persistent antigenic stimulation.16 

Neoplastic cells in MALT lymphomas exhibit features of MZ B cells from which they are thought to derive within the MALT.19,20,51-54  In MALT lymphomas, the MZ is expanded and surrounds residual GCs (Figure 4A). The neoplastic cells are small and resemble centrocytes, and hence are often called “centrocyte-like cells” (CCLs) (Figure 4B). They extend in the adjacent epithelial mucosa and invade the glandular epithelium, producing pathognomonic lympho-epithelial lesions (LELs) (Figure 4B-C).18  A prominent plasmacytic differentiation is common.

Figure 4.

Histopathologic illustration of MALT/MZ lymphomas. (A) Low-power view (× 50) of the lymphoid infiltration in a gastric biopsy sample tissue section stained with hematoxylin and eosin from a patient with H pylori–associated gastric MALT lymphoma. The arrow shows a germinal center surrounded by an enlarged marginal zone infiltrating the gastric lamina propria. (B) High-power view (× 400) of an intestinal biopsy sample tissue section stained with hematoxylin and eosin from a patient with C jejuni–associated IPSID, revealing centrocyte-like cells (CCLs) infiltrating the crypt epithelium and forming lymphoepithelial lesions (LELs). (C) Sections of jejunum (× 100) stained with primary antibodies directed against the B-cell marker CD20 (appears brown when stained with enzyme-linked secondary antibodies) and counterstained with hematoxylin show CD20+ centrocyte-like lymphocytes pervading the lamina propria surrounding crypts. (The top inset shows a higher power view of the epithelium.) CD20+ CCLs infiltrate the crypt epithelium and produce characteristic LELs (arrow). (D) High-power view (× 400) of a gastric biopsy sample tissue section stained according to the Giemsa technique from a patient with H pylori –associated gastric MALT lymphoma. The gastric mucosa is heavily infected by H pylori (arrow). (E) Immunohistochemical analysis of a jejunal section (× 400) from a patient with IPSID and stained with an anti–C jejuni monoclonal antibody (brown) and hematoxylin. The arrows point to immunolabeled material shown at a higher magnification in the top-right inset. The top-left inset shows a crypt section with intraluminal immunolabeled bacteria. (F) A blood smear (× 1000) colored according to the May-Grünwald-Giemsa technique showing a typical villous lymphocyte from a patient with HCV-associated splenic lymphoma with villous lymphocytes. Sections were viewed using a Zeiss Axiophot microscope (Carl Zeiss, Thornwood, NY) and Olympus UPlan F1 5 ×/0.12 numeric aperture (NA) (A), 40 ×/0.7 NA (B, D, E), 10 ×/0.30 (C), and 100 ×/0.30 NA (F; oil immersion) objectives (Olympus, Melville, NY), and were photographed using a Spot RT digital camera and Spot 3 software (both from Diagnostic Instruments, Sterling Heights, MI).

Figure 4.

Histopathologic illustration of MALT/MZ lymphomas. (A) Low-power view (× 50) of the lymphoid infiltration in a gastric biopsy sample tissue section stained with hematoxylin and eosin from a patient with H pylori–associated gastric MALT lymphoma. The arrow shows a germinal center surrounded by an enlarged marginal zone infiltrating the gastric lamina propria. (B) High-power view (× 400) of an intestinal biopsy sample tissue section stained with hematoxylin and eosin from a patient with C jejuni–associated IPSID, revealing centrocyte-like cells (CCLs) infiltrating the crypt epithelium and forming lymphoepithelial lesions (LELs). (C) Sections of jejunum (× 100) stained with primary antibodies directed against the B-cell marker CD20 (appears brown when stained with enzyme-linked secondary antibodies) and counterstained with hematoxylin show CD20+ centrocyte-like lymphocytes pervading the lamina propria surrounding crypts. (The top inset shows a higher power view of the epithelium.) CD20+ CCLs infiltrate the crypt epithelium and produce characteristic LELs (arrow). (D) High-power view (× 400) of a gastric biopsy sample tissue section stained according to the Giemsa technique from a patient with H pylori –associated gastric MALT lymphoma. The gastric mucosa is heavily infected by H pylori (arrow). (E) Immunohistochemical analysis of a jejunal section (× 400) from a patient with IPSID and stained with an anti–C jejuni monoclonal antibody (brown) and hematoxylin. The arrows point to immunolabeled material shown at a higher magnification in the top-right inset. The top-left inset shows a crypt section with intraluminal immunolabeled bacteria. (F) A blood smear (× 1000) colored according to the May-Grünwald-Giemsa technique showing a typical villous lymphocyte from a patient with HCV-associated splenic lymphoma with villous lymphocytes. Sections were viewed using a Zeiss Axiophot microscope (Carl Zeiss, Thornwood, NY) and Olympus UPlan F1 5 ×/0.12 numeric aperture (NA) (A), 40 ×/0.7 NA (B, D, E), 10 ×/0.30 (C), and 100 ×/0.30 NA (F; oil immersion) objectives (Olympus, Melville, NY), and were photographed using a Spot RT digital camera and Spot 3 software (both from Diagnostic Instruments, Sterling Heights, MI).

Close modal

Gastric MALT lymphoma and Helicobacter pylori: a model for infection-associated MALT lymphomas. Gastric MALT lymphoma is the most common MALT lymphoma and represents the majority of lymphomas involving the stomach.15,17,18  Gastric MALT lymphoma develops on a background of chronic inflammation and lymphoid infiltration displaying features of classic MALT architecture, of which the gastric mucosa is physiologically devoid.17 

Gastroduodenal Helicobacter pylori infection is strongly associated with gastric MALT lymphoma18  (Figure 4D). H pylori is a Gram-negative bacterium that colonizes the gastric mucosa and is associated with peptic ulcers and gastric adenocarcinomas.55-57 Hpylori is present in gastric biopsy samples of the majority of patients with gastric MALT lymphoma.58  The incidence of gastric MALT lymphoma is the highest in regions where H pylori infection is endemic,59  and the seroprevalence of H pylori is higher in patients with gastric MALT lymphomas60  than in control patients without MALT lymphomas. Eradication of H pylori leads to complete regression of the lymphoma during the early stages of the disease in nearly 80% of the cases.61-64  Low-grade gastric MALT lymphoma may evolve into aggressive large-cell gastric lymphoma, which is typically refractory to antibiotics.17,65-67  Retrospective analyses have shown that clonal B cells were already present in the gastritis, years before the clinical emergence of the lymphoma.68 

Immunoproliferative small intestinal disease: the quest for a microbial association. Immunoproliferative small intestinal disease (IPSID), also called alpha heavy-chain (αHC) disease or Mediterranean lymphoma, is a MALT lymphoma arising in the small intestine. The malignant cells have a distinctive lymphoplasmacytic phenotype and secrete a monotypic, truncated immunoglobulin α-heavy chain lacking an associated light chain, which can be detected as a paraprotein in the serum of patients.69-71  Histologic features of IPSID range from early lymphoplasmacytic intestinal infiltration to overt malignant diffuse large B-cell lymphoma. IPSID shares all the histologic features of MALT lymphomas, namely the presence of CCL, LEL, and plasma-cell differentiation.47,72  IPSID is chiefly observed in young adults originating from the Mediterranean basin, the Middle East, the Far East, and Africa.71  Antimicrobial therapy with tetracycline, ampicillin, or metronidazole is effective in early-stage IPSID,71  a finding that led the first investigators to hypothesize (years before H pylori was identified and associated with gastric MALT lymphoma) that a microbial species might play an etiologic role in IPSID development.71  Nevertheless, despite repeated attempts using classic culture-based approaches, the efforts made toward the identification of a microbial species associated with IPSID remained vain. Although H pylori was recently proposed to be involved in IPSID, this proposition was not corroborated in a subsequent retrospective analysis of more than 20 cases.73,74 

Applying an unbiased molecular approach previously used with success to identify the bacterial species associated with bacillary angiomatosis (Bartonella henselae)75  and Whipple disease (Tropheryma whipplei),76  we have demonstrated the presence of C jejuni–specific sequences in intestinal tissue samples of an IPSID patient with a spectacular response to antimicrobial therapy. These results were confirmed by in situ hybridization and immunohistochemistry (Figure 4E) on this index case and 4 of 6 archival additional cases.77  Identification of C jejuni in 5 of 7 patients with IPSID and the dramatic response observed in the index patient after microbial eradication make for a strong argument for the association of C jejuni with IPSID.78  However, association is not proof for causation, although it is usually the first step in proving the microbial etiology of a disease. To definitively demonstrate that C jejuni infection causes IPSID, the Koch postulate would need to be fulfilled (ie, Is C jejuni detectable in the host's intestine in early stages of the disease? Can C jejuni be cultivated from the diseased tissue? Can C jejuni trigger the disease in an animal model? Can C jejuni be isolated from the diseased animal?). Note that recent advances in molecular diagnostic tools have led to the restatement of new criteria for this postulate that take into account the putative uncultivability and host specificity of microbial species identified by molecular techniques.79,80  Demonstration of long-term C jejuni intestinal persistence is important for incriminating this bacterial species in IPSID development. The epidemiology of C jejuni in developing countries, in which IPSID is exclusively observed, sharply contrasts with that reported in developed countries. Up to 15% of asymptomatic children in developing countries carry Campylobacter organisms in their stools, whereas in developed countries, fecal Campylobacter is present in less than 0.5%.81  It is not known whether C jejuni can persistently colonize the small intestinal mucosa without concomitant detectable fecal shedding. Future studies need to focus on the extent of the asymptomatic and long-term intestinal carriage, its putative correlation with lamina propria lymphoid infiltration that might precede the emergence of lymphoma, and ultimately its causal relationship with IPSID. The absence of a functional B-cell receptor in IPSID raises the question as to how antigens might persistently stimulate these cells. In the early stages of infection, antigen-specific B cells in the lamina propria responding to microbial antigens (and possibly cross-reacting with autoantigens) would be stimulated and proliferate. During persistent stimulation, mutations would accumulate, leading to the selection of a clone having lost the ability to express a complete immunoglobulin and thus rendered insensitive to a negative feedback loop.71  According to this model, the subsequent proliferation would depend on survival factors (eg, BAFF) induced in the inflamed mucosa by persisting infection, as well as a proliferative advantage of the clone due to the loss of the negative regulation of normal IgA synthesis.82 

Extraintestinal MALT-type lymphomas may also be associated with chronic infections due to so-far-unidentified bacterial species. A similar approach to that used for IPSID could show promises in identifying such bacterial species.

Other bacteria-associated MALT lymphomas. The presence of Borrelia burgdorferi has been reported in primary cutaneous B-cell lymphoma (PCBCL) tissue.83-85  Despite contradictory reports from investigators from other geographic areas,86,87  there is significant support for the hypothesis that B burgdorferi infection might be associated with chronic antigen-driven lymphomagenesis in the skin, a tissue in which B burgdorferi is known to establish a persistent infection.85,88  Skin is the portal of entry of B burgdorferi and is the most commonly affected tissue in Lyme borreliosis.88  Late in the disease, lymphocytes may infiltrate the dermis and produce the characteristic borrelial “lymphocytoma.” B burgdorferi is present within the early skin lesions of erythema migrans and later in lymphocytoma, and its DNA can be readily amplified from biopsies of diseased skin. PCBCL is a rare entity that commonly displays the histologic features of MZ lymphoma.89  The incidence of PCBCL is higher in areas endemic for Lyme disease, and borrelial DNA has been amplified from skin biopsies.85,90  Regression of the lymphoma after antimicrobial therapy has been reported.84,91,92  Histologically, borrelial lymphocytoma can be difficult to distinguish from PCBCL, and has led to the improper term of “Borrelia-associated pseudolymphoma.”93  Having in mind the H pylori model of gastric MALT lymphomagenesis, the cutaneous manifestations of Lyme borreliosis could be viewed as a multistep progression from lymphocytoma to “pseudolymphoma” eventually leading to PCBCL.90,93  Evidence of B-cell monoclonality may help distinguish between the different stages of the disease,90,93  although it does not constitute a definitive proof of malignancy.

Recently, Chlamydia psittaci infection has also been associated with ocular adnexal MALT lymphomas.94,95  Adnexal MALT lymphomas have been described in the context of chronic conjunctivitis, and particularly “inclusion conjunctivitis,” which can be associated with Chlamydia infection.96,97  The presence of Chlamydia psittaci DNA in biopsy material and peripheral-blood mononuclear cells from patients with ocular adnexal lymphomas94  was demonstrated by targeted polymerase chain reaction (PCR), and C psittaci DNA was detected in 80% of ocular adnexal lymphoma samples. In some patients, antimicrobial treatment with doxycycline was associated with a clinical response.94  Together, these data argue for a putative role for C psittaci in ocular adnexal MALT lymphomagenesis.

Splenic marginal zone lymphoma (SMZL) and splenic lymphoma with villous lymphocytes (SLVL)

SMZL is a rare low-grade B-cell lymphoma involving predominantly the spleen.54  A leukemic phase with cytologically distinct lymphocytes defines the SLVL variant of SMZL.98  SMZL usually presents as an indolent lymphoma, and autoimmune manifestations such as serum rheumatoid factor (RF) are frequently associated. Histologically, the marginal zone surrounding the follicular areas is expanded and neoplastic cells have cytologic and phenotypical features of marginal zone lymphocytes,98,99  clearly distinguishing them from the lymphocytes present in the follicular center or the mantle area.

Hepatitis C virus (HCV) infection and MZ lymphomas: a correlation between lymphoid proliferation and viral load. We have reported the association of a subset of SLVL with chronic HCV infection,100,101  a finding that has now been confirmed by other investigators.102  HCV is an RNA virus associated with extrahepatic manifestations, such as essential mixed cryoglobulinemia (EMC) and B-cell lymphoproliferations.103  HCV-associated SLVL is indistinguishable from classic SLVL, except for the presence of HCV viral replication.100  EMC is consistently present in HCV-associated SLVL.101  Antiviral treatment with interferon alfa with or without ribavirin results in a marked reduction of lymphocytosis and splenomegaly in HCV-associated SLVL, whereas it is ineffective in HCV-negative SLVL.100,101  Complete virologic response correlates with sustained hematologic response, and virologic relapse is associated with re-emergence of circulating villous lymphocytes and splenomegaly. Reduction in HCV viral load after restarting the antiviral treatment correlates with hematologic remissions. Overall, these data indicate a strong correlation between serum viral load and tumor burden in HCV-associated SLVL, and support the existence of a causal relationship between HCV chronic antigenic stimulation and the MZ lymphomatous process.104 

Other HCV-associated lymphomas. EMC is considered a nonmalignant B-cell lymphoproliferation characterized by the synthesis of a monoclonal IgM with RF activity against immune complexes containing HCV proteins.105-107  Several epidemiologic studies have reported an association between HCV infection and B-cell lymphomas,108-112  and most cases of these HCV-associated lymphomas are low-grade MZ lymphomas.53,109,112  MALT lymphomas of the salivary glands may be associated with HCV infection.113  Monoclonal B cells can be detected during chronic HCV infection, especially in patients with HCV-associated ECM.103  Furthermore, the existence of a cryoglobulinemia is an independent risk factor for lymphomas in HCV-infected patients112  and may thus be considered as an early marker of HCV-associated lymphoproliferation.

MZ lymphomas and autoimmunity

Sjögren syndrome (SS) and autoimmune Hashimoto thyroiditis (HT) are characterized by autoreactive T- and B-lymphocyte infiltration of the salivary glands and the thyroid, respectively. Chronic inflammation and the ensuing cellular damage are associated with massive exposure of autoantigens to the immune system. B lymphocytes infiltrating the salivary glands in SS and the thyroid in HT progressively organize into a lymphoid infiltrate that reproduces the distinctive histologic architecture of normal MALT, including the presence of numerous reactive follicles.114  The risk for developing a B-cell lymphoma is increased by a factor of 44 in patients with SS.8  Similarly, indolent lymphomas of the thyroid, most often of the MALT-type, develop on a background of autoimmunity.115,116  Thus, utoantigenic stimulation observed during SS and AT appears to recapitulate the chronic microbial antigenic stimulation observed during persisting infections, and as in chronic infections, the failure to eradicate the antigenic source in autoimmunity leads to sustained B-cell stimulation, thus favoring lymphoid transformation and lymphomas.

H pylori and gastric MALT lymphoma

Converging evidence supports a causative role for H pylori in gastric MALT lymphomas117,118 : T cells from patients with gastric MALT lymphomas are able to sustain the in vitro proliferation of autologous malignant B cells in the presence of H pylori extracts, in a CD40-CD40L–dependent manner, supporting the role of H pylori in triggering the lymphoproliferation119  (Figure 1A). Strikingly, neoplastic B cells from gastric MALT lymphoma are not specific for H pylori antigens but rather for autoantigens found in the gastric mucosa. These autoreactive B cells are thought to receive cognate help from H pylori–specific T cells displaying cross-reactivity with gastric autoantigens (Figure 1A and “Microbial persistence: implications in lymphoproliferation and autoimmunity”). Thus, the malignant B cells could derive from TD GC B cells that homed to the MZ and thus display MZ-type phenotype and function. Alternatively, they could also be true MZ B cells participating in a TD response.28,30 

H pylori–infected gastric mucosal cells produce proinflammatory cytokines (such as lymphotoxin beta) and B-cell homing factors (such as BCA-1), leading to the emergence of MALT in the gastric mucosa.120  Infection of mice with Helicobacter species, including H pylori and the related species H felis and H heilmannii, also leads to the development of chronic gastritis and gastric MALT lymphomas with similarities to the human disease.117,118 

Thus H pylori not only fulfills the Koch postulate for gastric ulcer and carcinomas, but can also be convincingly incriminated in gastric MALT lymphomagenesis. H pylori–associated gastric MALT lymphomagenesis thus stands as the best-defined paradigm for infection-associated indirect lymphoid transformation.

Microbial persistence: implications in lymphoproliferation and autoimmunity

Pathogens inducing chronic infection have selected countless mechanisms allowing them to persist in the host and colonize their specific niches.

Molecular mimicry, a situation in which microbial pathogens express antigenic motifs shared with the host, is also a mechanism that favors microbial persistence, given the tolerization of the immune system toward autoantigens. Several H pylori antigens resemble autoantigens, notably the fucosylated Lewis antigens expressed on the surface of the gastric mucosa and the epitopes of self-gastric parietal-cell H(+)K(+)-ATPase57,121  (Figure 1A). Other examples of autoreactivity elicited by antimicrobial immune responses have been described for the aforementioned microbial species associated with antigen-driven lymphomagenesis: C jejuni is associated with Guillain-Barré syndrome, an acute polyneuropathy induced by cross-reactive antibodies directed against C jejuni lipo-oligosaccharides (LOSs) and nervous system gangliosides122 ; B burgdorferi OspA protein is structurally homologous to human lymphocyte function antigen-1 (LFA1), which may play a role in the autoimmune manifestations of the disease.123 Chlamydia species also share immunoreactivity with eukaryotic heat shock proteins,124  and this has been proposed to play a role in autoimmunity associated with this bacterium. Finally, the basis for the strong association between the immune response to HCV and the detection of an RF may lie in the structural and antigenic homologies between the N-terminal region of the HCV E2 envelope protein and the human immunoglobulin variable domains, and as such can be recognized by antihuman antibodies.125,126 

All the aforementioned microbial pathogens may also evade the immune system by antigenic variation, as has been documented in detail for H pylori,57 B burgdorferi,127  and HCV.128  This process also contributes to chronic stimulation of the immune system by continuously modifying microbial antigenic determinants.

Evidence for antigenic selection in MZ lymphomas

Indirect evidence for the role of an antigen in B-cell proliferation can be deduced from the analysis of the V gene use and SHM in immunoglobulin V genes, because they constitute molecular signatures for antigen selection.129-131  All MZ lymphomas associated with chronic infection and/or autoimmunity exhibit a biased immunoglobulin V gene use and SHM.132-134  The recent finding that MALT lymphomas stand out among other B-cell lymphomas as frequently expressing immunoglobulin V genes with strong homology to RF135  underscores the links between chronic antigenic stimulation, autoimmunity, and development of MZ-derived lymphoproliferations. Analysis of the immunoglobulin specificity from 2 HCV-associated lymphoma tumor cells demonstrated that they bound the HCV E2 glycoprotein similarly to human anti-E2 antibodies.125  Furthermore, B-cell clones in HCV-associated EMC and lymphomas often use the VH1-69 gene segment, which is also used by anti-E2 antibodies elicited by HCV.133,134,136  Many autoantigens have been identified in both SS and AT, and a common feature to both conditions is the frequent presence of RF; some cases of salivary gland lymphomas arising in SS also use immunoglobulin segments with RF activity, further supporting the role of chronic antigen stimulation in the pathogenesis of this condition.137 

Ongoing mutations in proliferating lymphocytes

Reactive oxygen species (ROSs) produced during inflammation are genotoxic and favor the occurrence of oncogenic DNA damage in proliferating lymphocytes.138  The intrinsic genetic instability of B cells during isotype class-switching and SHM139,140  also increases the risk of transformation during protracted proliferation associated with inflammation.

H pylori chronic infection is associated with the production of ROSs, chronic inflammation, and DNA damage.57,141,142  Most isolates of C jejuni produce a toxin called CdtB (cytolethal distending toxin B) that causes direct DNA damage.143  CdtB induces double-strand DNA breaks and growth arrest in T lymphocytes and may thus participate in immune evasion mechanisms during infection with CdtB-producing bacteria,143  as well as the emergence of the DNA breaks in B cells leading to the synthesis of a truncated immunoglobulin as seen in IPSID patients.71  The conjunction of CdtB and SHM occurring in the MALT during C jejuni chronic infection could lead to large deletions of the variable region of the αHC, precluding the association with a light chain and leading to the synthesis of a truncated αHC.139,143 

Mechanisms of clonal progression of antigen-dependent B cells

Inactivation of cell-cycle regulating genes such as the cyclin-dependent kinase inhibitors p15 and p16 is observed in early stages of gastric MALT lymphomas (Figure 1B).144  Fas/CD95, involved in apoptosis and homeostasis of normal and autoreactive B cells,145  is often mutated in nongastric MALT lymphomas and in other MZ lymphomas.146,147  These alterations confer a clonal advantage to antigen-specific B cells and ultimately lead to transformation. Early transformed B cells would still rely on signals from the antigen receptor for their proliferation and survival, as attested by their antigen dependence, which is illustrated by the efficacy of antigenic eradication (Figure 1B).

Molecular pathogenesis and cytogenetic features of MZ lymphomas

Recurrent cytogenetic abnormalities are found in most MZ/MALT lymphomas. The t(11;18), which fuses the API2 and MALT1 genes and generates a functional API2-MALT1 fusion product, has been found in several cases of MALT lymphomas arising in various mucosal sites.18  t(11;18) is usually the sole chromosomal aberration and occurs early. Other translocations, including t(1;14) and t(14;18), which fuse the BCL10 and MALT1 genes, respectively, to the IGH locus (Figures 1C, 5), have also been described, and can be associated with other cytogenetic aberrations such as chromosome 3 trisomy.18  More recently, the occurrence of a t(3;14) translocation has been described in 10% of MALT lymphomas, but not in nodal or splenic MZ lymphomas. This translocation that fuses FoxP1 to the IgH locus is mutually exclusive with the other MALT lymphoma–specific translocations, t(11;18), t(1;14), and t(14;18).148 

In SMZLs, the most common cytogenetic abnormality is the deletion of the long arm of chromosome 7 (7q21-32), which likely involves cdk6, and trisomy 3,54  whereas the t(11;18), t(1;14), and t(14;18) translocations are not found.

The oncogenic activity of the 3 chromosomal translocations t(11;18), t(1;14), and t(14;18) is linked to the physiologic role of BCL10 and MALT1 in antigen receptor–mediated NF-κB activation and inhibition of apoptosis.22,149,150  Constitutive activation of the NF-κB pathway by these translocations bypasses the requirement for the B-cell receptor signaling and accounts for the antigen independence of cells harboring these translocations67,151,152  (Figures 1 and 5). The oncogenic role of the IgH-FoxP1 fusion transcript is unknown.22 

Figure 5.

Gastric MALT lymphoma translocations lead to a bypass of the BCR-mediated NF-κB activation.

Figure 5.

Gastric MALT lymphoma translocations lead to a bypass of the BCR-mediated NF-κB activation.

Close modal

Alterations of B-cell functions by HCV

The HCV E2 glycoprotein interacts with CD81 on the surface of B lymphocytes and is a target of the humoral response against the virus.125,153  CD81 engagement on B cells enhances signaling through the BCR.154  Engagement of CD81 and virus-specific BCR by E2 could perturb B-cell function and lead to lymphoma. Mutations in the p53, Bcl6, and β-catenin genes may occur in B-cell lines infected in vitro with HCV as well as in peripheral-blood mononuclear cells from patients with chronic HCV infection. Induction of nitric oxide synthase by the viral core protein (C) and nonstructural protein 3 (NS3) has been implicated in the occurrence of these mutations.155,156 

Although HCV can infect B cells in vitro, possibly through CD81,153  only one case of B-cell lymphoma associated with direct infection of B cells by HCV has been described so far.157  A lymphoid cell line derived from an HCV-infected patient presenting with a mantle-cell lymphoma produced virus in vitro.158  Thus, direct infection of lymphocyte by HCV does not appear to be a prerequisite for most HCV-associated lymphomas,159  in agreement with what one would expect for an antigen-driven lymphoproliferation according to the “indirect” model of lymphomagenesis.160  Moreover, recombinant HCV E2 binding to CD81 on B cells has been shown to induce hypermutations at the immunoglobulin locus.161  E2 is exposed on the virion surface and can interact externally with the CD81 coreceptor on B cells. This mechanism of mutagenesis would be independent of direct infection of B cells by HCV.

Antigen-driven lymphoproliferations constitute a pathophysiologic concept that took root with the description of the MALT-lymphoma entity in the early 1980s and bloomed with the identification of H pylori as the causal agent of gastric MALT lymphoma in the early 1990s. This concept assumes that the marginal zone B lymphocyte is the cell from which these types of lymphomas derive, through a protracted proliferation induced by a persisting antigen, mostly of microbial origin. This entity now includes numerous additional examples of lymphoproliferations, which fall into the wider category of “MZ lymphomas.” The recent deciphering of the antigenic specificity of B-cell responses from the MZ has yielded important clues in relation to MZ lymphomagenesis associated with persistent antigenic stimulation.16  The spectacular responses observed after microbial eradication in a number of MZ/MALT lymphomas associated with chronic infections are of great pathophysiologic but also clinical importance, because many patients can be treated without antineoplastic chemotherapy, at least during the early stage of the disease. Unbiased approaches aimed at the identification of novel or unsuspected pathogens associated with MZ/MALT lymphomas will undoubtedly lead to the lengthening of the list of microbial pathogens associated with lymphoproliferations. Additional work is now critically needed to provide irrefutable evidence demonstrating not only the association but also the causative role of the microbial pathogens identified with this approach.

Prepublished online as Blood First Edition Paper, January 5, 2006; DOI 10.1182/blood-2005-09-3679.

Note added in proof. C psittaci association with adnexal lymphoma has not been confirmed by other groups,162  emphasizing the possible geographic heterogeneity of the association.

We thank Nicole Brousse, Antoine Martin, and Françoise Valensi for kindly providing pictures for Figure 4 and Bruno Varet for his support. We apologize to colleagues whose work could not be cited because of space limitations.

1
Hennessy BT, Hanrahan EO, Daly PA. Non-Hodgkin lymphoma: an update.
Lancet Oncol.
2004
;
5
:
341
-353.
2
Pagano JS, Blaser M, Buendia MA, et al. Infectious agents and cancer: criteria for a causal relation.
Semin Cancer Biol.
2004
;
14
:
453
-471.
3
Kuppers R, Klein U, Hansmann ML, Rajewsky K. Cellular origin of human B-cell lymphomas.
N Engl J Med.
1999
;
341
:
1520
-1529.
4
Pasqualucci L, Neumeister P, Goossens T, et al. Hypermutation of multiple proto-oncogenes in B-cell diffuse large-cell lymphomas.
Nature.
2001
;
412
:
341
-346.
5
Shaffer AL, Rosenwald A, Staudt LM. Lymphoid malignancies: the dark side of B-cell differentiation.
Nat Rev Immunol.
2002
;
2
:
920
-932.
6
Carbone A. Emerging pathways in the development of AIDS-related lymphomas.
Lancet Oncol.
2003
;
4
:
22
-29.
7
Bower M.
The management of lymphoma in the immunosuppressed patient: best practice & research clinical haematology.
2002
;
15
:
517
-532.
8
Royer B, Cazals-Hatem D, Sibilia J, et al. Lymphomas in patients with Sjogren's syndrome are marginal zone B-cell neoplasms, arise in diverse extranodal and nodal sites, and are not associated with viruses.
Blood.
1997
;
90
:
766
-775.
9
Voulgarelis M, Moutsopoulos HM. Lymphoproliferation in autoimmunity and Sjogren's syndrome.
Curr Rheumatol Rep.
2003
;
5
:
317
-323.
10
Harris NL, Jaffe ES, Diebold J, et al. World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting-Airlie House, Virginia, November 1997.
J Clin Oncol.
1999
;
17
:
3835
-3849.
11
Seto M. Genetic and epigenetic factors involved in B-cell lymphomagenesis.
Cancer Sci.
2004
;
95
:
704
-710.
12
Okano M. Haematological association of Epstein-Barr virus infection.
Baillières Clin Haematol.
2000
;
13
:
199
-214.
13
Boshoff C, Weiss R. AIDS-related malignancies.
Nat Rev Cancer.
2002
;
2
:
373
-382.
14
Bazarbachi A, Ghez D, Lepelletier Y, et al. New therapeutic approaches for adult T-cell leukaemia.
Lancet Oncol.
2004
;
5
:
664
-672.
15
Cavalli F, Isaacson PG, Gascoyne RD, Zucca E. MALT Lymphomas.
Hematology (Am Soc Hematol Educ Program).
2001
:
241
-258.
16
Morse HC III, Kearney JF, Isaacson PG, Carroll M, Fredrickson TN, Jaffe ES. Cells of the marginal zone: origins, function and neoplasia.
Leuk Res.
2001
;
25
:
169
-178.
17
Du M, Isaacson P. Gastric MALT lymphoma: from aetiology to treatment.
Lancet Oncol.
2002
;
3
:
97
-104.
18
Isaacson PG, Du MQ. MALT lymphoma: from morphology to molecules.
Nat Rev Cancer.
2004
;
4
:
644
-653.
19
de Wolf-Peeters C, Pittaluga S, Dierlamm J, Wlodarska I, Van Den Berghe H. Marginal zone B-cell lymphomas including mucosa-associated lymphoid tissue type lymphoma (MALT), monocytoid B-cell lymphoma and splenic marginal zone cell lymphoma and their relation to the reactive marginal zone.
Leuk Lymphoma.
1997
;
26
:
467
-478.
20
Berger F, Felman P, Thieblemont C, et al. Non-MALT marginal zone B-cell lymphomas: a description of clinical presentation and outcome in 124 patients.
Blood.
2000
;
95
:
1950
-1956.
21
Aljurf MD, Owaidah TW, Ezzat A, Ibrahim E, Tbakhi A. Antigen- and/or immune-driven lymphoproliferative disorders.
Ann Oncol.
2003
;
14
:
1595
-1606.
22
Farinha P, Gascoyne RD. Molecular pathogenesis of mucosa-associated lymphoid tissue lymphoma.
J Clin Oncol.
2005
;
23
:
6370
-6378.
23
Martin F, Oliver AM, Kearney JF. Marginal zone and B1 B cells unite in the early response against T-independent blood-borne particulate antigens.
Immunity.
2001
;
14
:
617
-629.
24
Pillai S, Cariappa A, Moran ST. Marginal zone B cells.
Annu Rev Immunol.
2005
;
23
:
161
-196.
25
Oliver AM, Martin F, Kearney JF. IgMhighCD21high lymphocytes enriched in the splenic marginal zone generate effector cells more rapidly than the bulk of follicular B cells.
J Immunol.
1999
;
162
:
7198
-7207.
26
Weller S, Braun MC, Tan BK, et al. Human blood IgM “memory” B cells are circulating splenic marginal zone B cells harboring a prediversified immunoglobulin repertoire.
Blood.
2004
;
104
:
3647
-3654.
27
Willenbrock K, Jungnickel B, Hansmann ML, Kuppers R. Human splenic marginal zone B cells lack expression of activation-induced cytidine deaminase.
Eur J Immunol.
2005
;
35
:
3002
-3007.
28
Martin F, Kearney JF. Marginal-zone B cells.
Nat Rev Immunol.
2002
;
2
:
323
-335.
29
Cariappa A, Pillai S. Antigen-dependent B-cell development.
Curr Opin Immunol.
2002
;
14
:
241
-249.
30
Weller S, Reynaud CA, Weill JC. Splenic marginal zone B cells in humans: where do they mutate their Ig receptor?
Eur J Immunol.
2005
;
35
:
2789
-2792.
31
Song H, Cerny J. Functional heterogeneity of marginal zone B cells revealed by their ability to generate both early antibody-forming cells and germinal centers with hypermutation and memory in response to a T-dependent antigen.
J Exp Med.
2003
;
198
:
1923
-1935.
32
Wen L, Brill-Dashoff J, Shinton SA, Asano M, Hardy RR, Hayakawa K. Evidence of marginalzone B cell-positive selection in spleen.
Immunity.
2005
;
23
:
297
-308.
33
He B, Qiao X, Cerutti A. CpG DNA induces IgG class switch DNA recombination by activating human B cells through an innate pathway that requires TLR9 and cooperates with IL-10.
J Immunol.
2004
;
173
:
4479
-4491.
34
Kuppers R. B cells under influence: transformation of B cells by Epstein-Barr virus.
Nat Rev Immunol.
2003
;
3
:
801
-812.
35
Thorley-Lawson DA, Gross A. Persistence of the Epstein-Barr virus and the origins of associated lymphomas.
N Engl J Med.
2004
;
350
:
1328
-1337.
36
Thorley-Lawson DA. Epstein-Barr virus: exploiting the immune system.
Nat Rev Immunol.
2001
;
1
:
75
-82.
37
Quezada SA, Jarvinen LZ, Lind EF, Noelle RJ. CD40/CD154 interactions at the interface of tolerance and immunity.
Annu Rev Immunol.
2004
;
22
:
307
-328.
38
Kilger E, Kieser A, Baumann M, Hammerschmidt W. Epstein-Barr virus-mediated B-cell proliferation is dependent upon latent membrane protein 1, which simulates an activated CD40 receptor.
EMBO J.
1998
;
17
:
1700
-1709.
39
Dirmeier U, Neuhierl B, Kilger E, Reisbach G, Sandberg ML, Hammerschmidt W. Latent membrane protein 1 is critical for efficient growth transformation of human B cells by Epstein-Barr virus.
Cancer Res.
2003
;
63
:
2982
-2989.
40
Caldwell RG, Wilson JB, Anderson SJ, Longnecker R. Epstein-Barr virus LMP2A drives B cell development and survival in the absence of normal B cell receptor signals.
Immunity.
1998
;
9
:
405
-411.
41
Damania B. Oncogenic gamma-herpesviruses: comparison of viral proteins involved in tumorigenesis.
Nat Rev Microbiol.
2004
;
2
:
656
-668.
42
Casola S, Otipoby KL, Alimzhanov M, et al. B cell receptor signal strength determines B cell fate.
Nat Immunol.
2004
;
5
:
317
-327.
43
Hengge UR, Ruzicka T, Tyring SK, et al. Update on Kaposi's sarcoma and other HHV8 associated diseases: part 2, pathogenesis, Castleman's disease, and pleural effusion lymphoma.
Lancet Infect Dis.
2002
;
2
:
344
-352.
44
Manns A, Hisada M, La Grenade L. Human T-lymphotropic virus type I infection.
Lancet.
1999
;
353
:
1951
-1958.
45
Kurokawa M, Ghosh SK, Ramos JC, et al. Azidothymidine inhibits NF-kappaB and induces Epstein-Barr virus gene expression in Burkitt lymphoma.
Blood.
2005
;
106
:
235
-240.
46
Harris NL, Jaffe ES, Stein H, et al. A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group.
Blood.
1994
;
84
:
1361
-1392.
47
Isaacson P, Wright DH. Malignant lymphoma of mucosa-associated lymphoid tissue: a distinctive type of B-cell lymphoma.
Cancer.
1983
;
52
:
1410
-1416.
48
Isaacson P, Wright DH. Extranodal malignant lymphoma arising from mucosa-associated lymphoid tissue.
Cancer.
1984
;
53
:
2515
-2524.
49
Zucca E, Bertoni F, Roggero E, Cavalli F. The gastric marginal zone B-cell lymphoma of MALT type.
Blood.
2000
;
96
:
410
-419.
50
Wotherspoon AC. Extragastric MALT lymphoma.
Gut.
2002
;
51
:
148
-149.
51
Spencer J, Perry ME, Dunn-Walters DK. Human marginal-zone B cells.
Immunol Today.
1998
;
19
:
421
-426.
52
Nathwani BN, Anderson JR, Armitage JO, et al. Marginal zone B-cell lymphoma: a clinical comparison of nodal and mucosa-associated lymphoid tissue types: Non-Hodgkin's Lymphoma Classification Project.
J Clin Oncol.
1999
;
17
:
2486
-2492.
53
Arcaini L, Paulli M, Boveri E, et al. Splenic and nodal marginal zone lymphomas are indolent disorders at high hepatitis C virus seroprevalence with distinct presenting features but similar morphologic and phenotypic profiles.
Cancer.
2004
;
100
:
107
-115.
54
Thieblemont C, Felman P, Callet-Bauchu E, et al. Splenic marginal-zone lymphoma: a distinct clinical and pathological entity.
Lancet Oncol.
2003
;
4
:
95
-103.
55
Parsonnet J, Friedman GD, Vandersteen DP, et al. Helicobacter pylori infection and the risk of gastric carcinoma.
N Engl J Med.
1991
;
325
:
1127
-1131.
56
Wotherspoon AC. Gastric lymphoma, carcinoma and Helicobacter pylori.
J R Soc Med.
1995
;
88
:
302
-303.
57
Blaser MJ, Atherton JC. Helicobacter pylori persistence: biology and disease.
J Clin Invest.
2004
;
113
:
321
-333.
58
Wotherspoon AC, Ortiz-Hidalgo C, Falzon MR, Isaacson PG. Helicobacter pylori-associated gastritis and primary B-cell gastric lymphoma.
Lancet.
1991
;
338
:
1175
-1176.
59
Doglioni C, Wotherspoon AC, Moschini A, de Boni M, Isaacson PG. High incidence of primary gastric lymphoma in northeastern Italy.
Lancet.
1992
;
339
:
834
-835.
60
Parsonnet J, Hansen S, Rodriguez L, et al. Helicobacter pylori infection and gastric lymphoma.
N Engl J Med.
1994
;
330
:
1267
-1271.
61
Wotherspoon AC, Doglioni C, Diss TC, et al. Regression of primary low-grade B-cell gastric lymphoma of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori.
Lancet.
1993
;
342
:
575
-577.
62
Wotherspoon AC, Doglioni C, de Boni M, Spencer J, Isaacson PG. Antibiotic treatment for low-grade gastric MALT lymphoma [letter].
Lancet.
1994
;
343
:
1503
.
63
Bayerdorffer E, Neubauer A, Rudolph B, et al. Regression of primary gastric lymphoma of mucosa-associated lymphoid tissue type after cure of Helicobacter pylori infection: MALT Lymphoma Study Group.
Lancet.
1995
;
345
:
1591
-1594.
64
Roggero E, Zucca E, Pinotti G, et al. Eradication of Helicobacter pylori infection in primary low-grade gastric lymphoma of mucosa-associated lymphoid tissue.
Ann Intern Med.
1995
;
122
:
767
-769.
65
Wotherspoon AC, Diss TC, Pan L, Singh N, Whelan J, Isaacson PG. Low grade gastric B-cell lymphoma of mucosa associated lymphoid tissue in immunocompromised patients.
Histopathology.
1996
;
28
:
129
-134.
66
Liu H, Ruskon-Fourmestraux A, Lavergne-Slove A, et al. Resistance of t(11;18) positive gastric mucosa-associated lymphoid tissue lymphoma to Helicobacter pylori eradication therapy.
Lancet.
2001
;
357
:
39
-40.
67
Liu H, Ye H, Dogan A, et al. T(11;18)(q21;q21) is associated with advanced mucosa-associated lymphoid tissue lymphoma that expresses nuclear BCL10.
Blood.
2001
;
98
:
1182
-1187.
68
Zucca E, Bertoni F, Roggero E, 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
;
338
:
804
-810.
69
Seligmann M, Danon F, Hurez D, Mihaesco E, Preud'homme JL. Alpha-chain disease: a new immunoglobulin abnormality.
Science.
1968
;
162
:
1396
-1397.
70
Rambaud JC, Bognel C, Prost A, et al. Clinicopathological study of a patient with “Mediterranean” type of abdominal lymphoma and a new type of IgA abnormality (“alpha chain disease”).
Digestion.
1968
;
1
:
321
-336.
71
Al-Saleem T, Al-Mondhiry H. Immunoproliferative small intestinal disease (IPSID): a model for mature B-cell neoplasms.
Blood.
2005
;
105
:
2274
-2280.
72
Isaacson PG, Dogan A, Price SK, Spencer J. Immunoproliferative small-intestinal disease: an immunohistochemical study.
Am J Surg Pathol.
1989
;
13
:
1023
-1033.
73
Fischbach W, Tacke W, Greiner A, Muller-Hermelink HK. Regression of immunoproliferative small intestinal disease after eradication of Helicobacter pylori.
Lancet.
1997
;
349
:
31
-32.
74
Malekzadeh R, Kaviani MJ, Tabei SZ, Abdolhadi B, Haghshenas M, Navab F. Lack of association between Helicobacter pylori infection and immunoproliferative small intestinal disease.
Arch Iran Med.
1999
;
2
:
1
-4.
75
Relman DA, Loutit JS, Schmidt TM, Falkow S, Tompkins LS. The agent of bacillary angiomatosis: an approach to the identification of uncultured pathogens.
N Engl J Med.
1990
;
323
:
1573
-1580.
76
Relman DA, Schmidt TM, MacDermott RP, Falkow S. Identification of the uncultured bacillus of Whipple's disease.
N Engl J Med.
1992
;
327
:
293
-301.
77
Lecuit M, Abachin E, Martin A, et al. Immunoproliferative small intestinal disease associated with Campylobacter jejuni.
N Engl J Med.
2004
;
350
:
239
-248.
78
Parsonnet J, Isaacson PG. Bacterial infection and MALT lymphoma.
N Engl J Med.
2004
;
350
:
213
-215.
79
Fredericks DN, Relman DA. Sequence-based identification of microbial pathogens: a reconsideration of Koch's postulates.
Clin Microbiol Rev.
1996
;
9
:
18
-33.
80
Franco EL, Correa P, Santella RM, Wu X, Goodman SN, Petersen GM. Role and limitations of epidemiology in establishing a causal association.
Semin Cancer Biol.
2004
;
14
:
413
-426.
81
Coker AO, Isokpehi RD, Thomas BN, Amisu KO, Obi CL. Human campylobacteriosis in developing countries.
Emerg Infect Dis.
2002
;
8
:
237
-244.
82
Uren TK, Johansen FE, Wijburg OL, Koentgen F, Brandtzaeg P, Strugnell RA. Role of the polymeric Ig receptor in mucosal B cell homeostasis.
J Immunol.
2003
;
170
:
2531
-2539.
83
Garbe C, Stein H, Dienemann D, Orfanos CE. Borrelia burgdorferi-associated cutaneous B cell lymphoma: clinical and immunohistologic characterization of four cases.
J Am Acad Dermatol.
1991
;
24
:
584
-590.
84
Cerroni L, Zochling N, Putz B, Kerl H. Infection by Borrelia burgdorferi and cutaneous B-cell lymphoma.
J Cutan Pathol.
1997
;
24
:
457
-461.
85
Goodlad JR, Davidson MM, Hollowood K, et al. Primary cutaneous B-cell lymphoma and Borrelia burgdorferi infection in patients from the Highlands of Scotland.
Am J Surg Pathol.
2000
;
24
:
1279
-1285.
86
Wood GS, Kamath NV, Guitart J, et al. Absence of Borrelia burgdorferi DNA in cutaneous B-cell lymphomas from the United States.
J Cutan Pathol.
2001
;
28
:
502
-507.
87
Li C, Inagaki H, Kuo TT, Hu S, Okabe M, Eimoto T. Primary cutaneous marginal zone B-cell lymphoma: a molecular and clinicopathologic study of 24 Asian cases.
Am J Surg Pathol.
2003
;
27
:
1061
-1069.
88
Stanek G, Strle F. Lyme borreliosis.
Lancet.
2003
;
362
:
1639
-1647.
89
Gronbaek K, Moller PH, Nedergaard T, et al. Primary cutaneous B-cell lymphoma: a clinical, histological, phenotypic and genotypic study of 21 cases.
Br J Dermatol.
2000
;
142
:
913
-923.
90
Colli C, Leinweber B, Mullegger R, Chott A, Kerl H, Cerroni L. Borrelia burgdorferi-associated lymphocytoma cutis: clinicopathologic, immunophenotypic, and molecular study of 106 cases.
J Cutan Pathol.
2004
;
31
:
232
-240.
91
Kutting B, Bonsmann G, Metze D, Luger TA, Cerroni L. Borrelia burgdorferi-associated primary cutaneous B cell lymphoma: complete clearing of skin lesions after antibiotic pulse therapy or intralesional injection of interferon alfa-2a.
J Am Acad Dermatol.
1997
;
36
:
311
-314.
92
Roggero E, Zucca E, Mainetti C, et al. Eradication of Borrelia burgdorferi infection in primary marginal zone B-cell lymphoma of the skin.
Hum Pathol.
2000
;
31
:
263
-268.
93
Grange F, Wechsler J, Guillaume JC, et al. Borrelia burgdorferi-associated lymphocytoma cutis simulating a primary cutaneous large B-cell lymphoma.
J Am Acad Dermatol.
2002
;
47
:
530
-534
94
Ferreri AJ, Guidoboni M, Ponzoni M, et al. Evidence for an association between Chlamydia psittaci and ocular adnexal lymphomas.
J Natl Cancer Inst.
2004
;
96
:
586
-594.
95
Cahill M, Barnes C, Moriarty P, Daly P, Kennedy S. Ocular adnexal lymphoma-comparison of MALT lymphoma with other histological types.
Br J Ophthalmol.
1999
;
83
:
742
-747.
96
Tovilla-Canales JL, Tovilla y Pomar JL, Ceron JR. Lymphoproliferative disorders of the ocular adnexa.
Curr Opin Ophthalmol.
2004
;
15
:
401
-405.
97
Yeung L, Tsao YP, Chen PY, Kuo TT, Lai LJ. Combination of adult inclusion conjunctivitis and mucosa-associated lymphoid tissue (MALT) lymphoma in a young adult.
Cornea.
2004
;
23
:
71
-75.
98
Troussard X, Valensi F, Duchayne E, et al. Splenic lymphoma with villous lymphocytes: clinical presentation, biology and prognostic factors in a series of 100 patients: Groupe Francais d'Hematologie Cellulaire (GFHC).
Br J Haematol.
1996
;
93
:
731
-736.
99
Isaacson PG. Splenic marginal zone lymphoma.
Blood.
1996
;
88
:
751
-752.
100
Hermine O, Lefrere F, Bronowicki JP, et al. Regression of splenic lymphoma with villous lymphocytes after treatment of hepatitis C virus infection.
N Engl J Med.
2002
;
347
:
89
-94.
101
Saadoun D, Suarez F, Lefrere F, et al. Splenic lymphoma with villous lymphocytes, associated with type II cryoglobulinemia and HCV infection: a new entity?
Blood.
2005
;
105
:
74
-76.
102
Kelaidi C, Rollot F, Park S, et al. Response to antiviral treatment in hepatitis C virus-associated marginal zone lymphomas.
Leukemia.
2004
;
18
:
1711
-1716.
103
Mazzaro C, Franzin F, Tulissi P, et al. Regression of monoclonal B-cell expansion in patients affected by mixed cryoglobulinemia responsive to alpha-interferon therapy.
Cancer.
1996
;
77
:
2604
-2613.
104
Vallisa D, Bernuzzi P, Arcaini L, et al. Role of anti-hepatitis C virus (HCV) treatment in HCV-related, low-grade, B-cell, non-Hodgkin's lymphoma: a multicenter Italian experience.
J Clin Oncol.
2005
;
23
:
468
-473.
105
Zignego AL, Brechot C. Extrahepatic manifestations of HCV infection: facts and controversies.
J Hepatol.
1999
;
31
:
369
-376.
106
Cacoub P, Costedoat-Chalumeau N, Lidove O, Alric L. Cryoglobulinemia vasculitis.
Curr Opin Rheumatol.
2002
;
14
:
29
-35.
107
Ferri C, Sebastiani M, Giuggioli D, et al. Mixed cryoglobulinemia: demographic, clinical, and serologic features and survival in 231 patients.
Semin Arthritis Rheum.
2004
;
33
:
355
-374.
108
Mazzaro C, Zagonel V, Monfardini S, et al. Hepatitis C virus and non-Hodgkin's lymphomas.
Br J Haematol.
1996
;
94
:
544
-550.
109
Zuckerman E, Zuckerman T, Levine AM, et al. Hepatitis C infection in patients with B-cell non-Hodgkin lymphoma.
Ann Intern Med.
1997
;
127
:
423
-438.
110
Luppi M, Longo G, Ferrari MG, et al. Clinicopathological characterization of hepatitis C virus-related B-cell non-Hodgkin's lymphomas without symptomatic cryoglobulinemia.
Ann Oncol.
1998
;
9
:
495
-498.
111
Talamani R, Montella M, Crovatto M, et al. Non-Hodgkin's lymphoma and hepatitis C virus: a case-control study from Northern and Southern Italy.
Int J Cancer.
2004
;
110
:
380
-385.
112
Monti G, Pioltelli P, Saccardo F, et al. Incidence and characteristics of non-Hodgkin lymphomas in a multicenter case file of patients with hepatitis C virus-related symptomatic mixed cryoglobulinemias.
Arch Intern Med.
2005
;
165
:
101
-105.
113
Ambrosetti A, Zanotti R, Pattaro C, 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
;
126
:
43
-49.
114
Hsi ED, Singleton TP, Svoboda SM, Schnitzer B, Ross CW. Characterization of the lymphoid infiltrate in Hashimoto thyroiditis by immunohistochemistry and polymerase chain reaction for immunoglobulin heavy chain gene rearrangement.
Am J Clin Pathol.
1998
;
110
:
327
-333.
115
Derringer GA, Thompson LD, Frommelt RA, Bijwaard KE, Heffess CS, Abbondanzo SL. Malignant lymphoma of the thyroid gland: a clinicopathologic study of 108 cases.
Am J Surg Pathol.
2000
;
24
:
623
-639.
116
Takakuwa T, Dong Z, Takayama H, Matsuzuka F, Nagata S, Aosaza K. Frequent mutations of Fas gene in thyroid lymphoma.
Cancer Res.
2001
;
61
:
1382
-1385.
117
Mueller A, O'Rourke J, Grimm J, et al. Distinct gene expression profiles characterize the histopathological stages of disease in Helicobacter-induced mucosa-associated lymphoid tissue lymphoma.
Proc Natl Acad Sci U S A.
2003
;
100
:
1292
-1297.
118
Lee A, O'Rourke J, Enno A. Gastric mucosa associated lymphoid tissue lymphoma: implications of animal models on pathogenesis and therapeutic considerations: mouse model of gastric lymphoma.
Rec Res Cancer Res.
2000
;
156
:
42
-51.
119
Hussell T, Isaacson PG, Crabtree JE, Spencer J. The response of cells from low-grade B-cell gastric lymphomas of mucosa-associated lymphoid tissue to Helicobacter pylori.
Lancet.
1993
;
342
:
571
-574.
120
Mazzucchelli L, Blaser A, Kappeler A, et al. BCA-1 is highly expressed in Helicobacter pylori-induced mucosa-associated lymphoid tissue and gastric lymphoma.
J Clin Invest.
1999
;
104
:
R49
-R54.
121
D'Elios MM, Appelmelk BJ, Amedei A, Bergman MP, Del Prete G. Gastric autoimmunity: the role of Helicobacter pylori and molecular mimicry.
Trends Mol Med.
2004
;
10
:
316
-323.
122
Yuki N. Molecular mimicry between gangliosides and lipopolysaccharides of Campylobacter jejuni isolated from patients with Guillain-Barre syndrome and Miller Fisher syndrome.
J Infect Dis.
1997
;
176
(suppl 2):
S150
-S153.
123
Gross DM, Forsthuber T, Tary-Lehmann M, et al. Identification of LFA-1 as a candidate autoantigen in treatment-resistant Lyme arthritis.
Science.
1998
;
281
:
703
-706.
124
Lamb DJ, El-Sankary W, Ferns GA. Molecular mimicry in atherosclerosis: a role for heat shock proteins in immunisation.
Atherosclerosis.
2003
;
167
:
177
-185.
125
Quinn ER, Chan CH, Hadlock KG, Foung SK, Flint M, Levy S. The B-cell receptor of a hepatitis C virus (HCV)-associated non-Hodgkin lymphoma binds the viral E2 envelope protein, implicating HCV in lymphomagenesis.
Blood.
2001
;
98
:
3745
-3749.
126
Hu YW, Rocheleau L, Larke B, et al. Immunoglobulin mimicry by hepatitis C virus envelope protein E2.
Virology.
2005
;
332
:
538
-549.
127
Singh SK, Girschick HJ. Molecular survival strategies of the Lyme disease spirochete Borrelia burgdorferi.
Lancet Infect Dis.
2004
;
4
:
575
-583.
128
Timm J, Lauer GM, Kavanagh DG, et al. CD8 epitope escape and reversion in acute HCV infection.
J Exp Med.
2004
;
200
:
1593
-1604.
129
Li Z, Woo CJ, Iglesias-Ussel MD, Ronai D, Scharff MD. The generation of antibody diversity through somatic hypermutation and class switch recombination.
Genes Dev.
2004
;
18
:
1
-11.
130
Dunn-Walters DK, Boursier L, Spencer J, Isaacson PG. Analysis of immunoglobulin genes in splenic marginal zone lymphoma suggests ongoing mutation.
Hum Pathol.
1998
;
29
:
585
-593.
131
Tierens A, Delabie J, Michiels L, Vandenberghe P, De Wolf-Peeters C. Marginal-zone B cells in the human lymph node and spleen show somatic hypermutations and display clonal expansion.
Blood.
1999
;
93
:
226
-234.
132
Du M, Diss TC, Xu C, Peng H, Isaacson PG, Pan L. Ongoing mutation in MALT lymphoma immunoglobulin gene suggests that antigen stimulation plays a role in the clonal expansion.
Leukemia.
1996
;
10
:
1190
-1197.
133
De Re V, De Vita S, Marzotto A, et al. Sequence analysis of the immunoglobulin antigen receptor of hepatitis C virus-associated non-Hodgkin lymphomas suggests that the malignant cells are derived from the rheumatoid factor-producing cells that occur mainly in type II cryoglobulinemia.
Blood.
2000
;
96
:
3578
-3584.
134
Marasca R, Vaccari P, Luppi M, et al. Immunoglobulin gene mutations and frequent use of VH1–69 and VH4–34 segments in hepatitis C virus-positive and hepatitis C virus-negative nodal marginal zone B-cell lymphoma.
Am J Pathol.
2001
;
159
:
253
-261.
135
Bende RJ, Aarts WM, Riedl RG, de Jong D, Pals ST, van Noesel CJ. Among B cell non-Hodgkin's lymphomas, MALT lymphomas express a unique antibody repertoire with frequent rheumatoid factor reactivity.
J Exp Med.
2005
;
201
:
1229
-1241.
136
De Re V, De Vita S, Gasparotto D, et al. Salivary gland B cell lymphoproliferative disorders in Sjogren's syndrome present a restricted use of antigen receptor gene segments similar to those used by hepatitis C virus-associated non-Hodgkins's lymphomas.
Eur J Immunol.
2002
;
32
:
903
-910.
137
Mariette X. Lymphomas complicating Sjogren's syndrome and hepatitis C virus infection may share a common pathogenesis: chronic stimulation of rheumatoid factor B cells.
Ann Rheum Dis.
2001
;
60
:
1007
-1010.
138
Coussens LM, Werb Z. Inflammation and cancer.
Nature.
2002
;
420
:
860
-867.
139
Goossens T, Klein U, Kuppers R. Frequent occurrence of deletions and duplications during somatic hypermutation: implications for oncogene translocations and heavy chain disease.
Proc Natl Acad Sci U S A.
1998
;
95
:
2463
-2468.
140
Greeve J, Philipsen A, Krause K, et al. Expression of activation-induced cytidine deaminase in human B-cell non-Hodgkin lymphomas.
Blood.
2003
;
101
:
3574
-3580.
141
Smoot DT, Elliott TB, Verspaget HW, et al. Influence of Helicobacter pylori on reactive oxygen-induced gastric epithelial cell injury.
Carcinogenesis.
2000
;
21
:
2091
-2095.
142
Arabski M, Klupinska G, Chojnacki J, et al. DNA damage and repair in Helicobacter pylori-infected gastric mucosa cells.
Mutat Res.
2005
;
570
:
129
-135.
143
Lara-Tejero M, Galan JE. A bacterial toxin that controls cell cycle progression as a deoxyribonuclease I-like protein.
Science.
2000
;
290
:
354
-357.
144
Martinez-Delgado B, Fernandez-Piqueras J, Garcia MJ, et al. Hypermethylation of a 5′ CpG island of p16 is a frequent event in non-Hodgkin's lymphoma.
Leukemia.
1997
;
11
:
425
-428.
145
Rathmell JC, Thompson CB. Pathways of apoptosis in lymphocyte development, homeostasis, and disease.
Cell.
2002
;
109
(suppl):
S97
-S107.
146
Gronbaek K, Straten PT, Ralfkiaer E, et al. Somatic Fas mutations in non-Hodgkin's lymphoma: association with extranodal disease and autoimmunity.
Blood.
1998
;
92
:
3018
-3024.
147
Gronbaek K, Ralfkiaer E, Kalla J, Skovgaard GL, Guldberg P. Infrequent somatic Fas mutations but no evidence of Bcl10 mutations or t(11;18) in primary cutaneous MALT-type lymphoma.
J Pathol.
2003
;
201
:
134
-140.
148
Streubel B, Vinatzer U, Lamprecht A, Raderer M, Chott A. T(3;14)(p14.1;q32) involving IGH and FOXP1 is a novel recurrent chromosomal aberration in MALT lymphoma.
Leukemia.
2005
;
19
:
652
-658.
149
Sanchez-Beato M, Sanchez-Aguilera A, Piris MA. Cell cycle deregulation in B-cell lymphomas.
Blood.
2003
;
101
:
1220
-1235.
150
Stoffel A, Chaurushiya M, Singh B, Levine AJ. Activation of NF-kappaB and inhibition of p53-mediated apoptosis by API2/mucosa-associated lymphoid tissue 1 fusions promote oncogenesis.
Proc Natl Acad Sci U S A.
2004
;
101
:
9079
-9084.
151
Liu H, Ye H, Ruskone-Fourmestraux A, et al. T(11;18) is a marker for all stage gastric MALT lymphomas that will not respond to H. pylori eradication.
Gastroenterology.
2002
;
122
:
1286
-1294.
152
Yeh KH, Kuo SH, Chen LT, et al. Nuclear expression of BCL10 or nuclear factor kappa B helps predict Helicobacter pylori-independent status of low-grade gastric mucosa-associated lymphoid tissue lymphomas with or without t(11;18)(q21; q21).
Blood.
2005
;
106
:
1037
-1041.
153
Pileri P, Uematsu Y, Campagnoli S, et al. Binding of hepatitis C virus to CD81.
Science.
1998
;
282
:
938
-941.
154
Fearon DT, Carroll MC. Regulation of B lymphocyte responses to foreign and self-antigens by the CD19/CD21 complex.
Annu Rev Immunol.
2000
;
18
:
393
-422.
155
Machida K, Cheng KT, Sung VM, et al. Hepatitis C virus induces a mutator phenotype: enhanced mutations of immunoglobulin and protooncogenes.
Proc Natl Acad Sci U S A.
2004
;
101
:
4262
-4267.
156
Machida K, Cheng KT, Sung VM, Lee KJ, Levine AM, Lai MM. Hepatitis C virus infection activates the immunologic (type II) isoform of nitric oxide synthase and thereby enhances DNA damage and mutations of cellular genes.
J Virol.
2004
;
78
:
8835
-8843.
157
Levine AM, Shimodaira S, Lai MM. Treatment of HCV-related mantle-cell lymphoma with ribavirin and pegylated interferon Alfa.
N Engl J Med.
2003
;
349
:
2078
-2079.
158
Sung VM, Shimodaira S, Doughty AL, et al. Establishment of B-cell lymphoma cell lines persistently infected with hepatitis C virus in vivo and in vitro: the apoptotic effects of virus infection.
J Virol.
2003
;
77
:
2134
-2146.
159
De Vita S, De Re V, Sansonno D, et al. Lack of HCV infection in malignant cells refutes the hypothesis of a direct transforming action of the virus in the pathogenesis of HCV-associated B-cell NHLs.
Tumori.
2002
;
88
:
400
-406.
160
Zignego AL, Ferri C, Giannini C, et al. Hepatitis C virus infection in mixed cryoglobulinemia and B-cell non-Hodgkin's lymphoma: evidence for a pathogenetic role.
Arch Virol.
1997
;
142
:
545
-555.
161
Machida K, Cheng KT, Pavio N, Sung VM, Lai MM. Hepatitis C virus E2-CD81 interaction induces hypermutation of the immunoglobulin gene in B cells.
J Virol.
2005
;
79
:
8079
-8089.
162
Rosado MF, Byrne GE Jr, Ding F, et al. Ocular adnexal lymphoma: a clinicopathologic study of a large cohort of patients with no evidence for an association with Chlamydia psittaci.
Blood.
2006
;
107
:
467
-472.
Sign in via your Institution