Molecular and clinical progress in follicular lymphoma lacking the t(14;18) translocation (Review)

  • Authors:
    • Zunmin Zhu
    • Tao Li
    • Xuran Zhang
    • Zhengqiang Zhang
    • Dandan Zhu
    • Pei Lin
    • Shichun Tu
    • Weihong Ren
  • View Affiliations

  • Published online on: November 21, 2019     https://doi.org/10.3892/ijo.2019.4917
  • Pages: 7-17
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Although the majority of patients with follicular lymphoma (FL) harbor the t(14;18)(q32;q21) IGH/BCL2 gene rearrangement that leads to the overexpression of BCL2 protein, approximately 20% of FL cases lack t(14;18)(q32;q21). It is considered that BCL2 overexpression underscores the development of the majority of cases of FL and their transformation to more aggressive lymphoma [known as transformed FL (tFL)]. However, FL cases lacking the t(14;18)(q32;q21) translocation exhibit symptoms analogous to their t(14;18)‑positive counterparts. An important goal of recent research on FL has been to clarify the distinctions between the two different forms of FL. Numerous studies have shed light onto the genetic and molecular features of t(14;18)‑negative FL and the related clinical manifestations. In this review, we summarize the current knowledge of t(14;18)‑negative FL occurring in the lymph nodes with an emphasis on the underlying molecular and clinical features. In addition, novel treatment directions are discussed.

1. Introduction

Follicular lymphoma (FL) is one of the most common hematological neoplasms. It is an indolent type of non-Hodgkin lymphoma that typically grows and spreads at a slow rate. In normal B cell development, resting follicular B cells are exposed to antigens and may eventually develop into memory B cells (Fig. 1A); in FL, neoplastic germinal center B cells accumulate to form neoplastic follicles (Fig. 1B). These follicles are usually composed of a mixture of cleaved centrocytes and noncleaved centroblasts (1,2). FL is the second most common form of nodal lymphoma, following diffuse large B-cell lymphoma (DLBCL) (3). It is diagnosed at a median age of 60 years, with life expectancy ranging from 6 to 10 years following initial diagnosis (3-5). However, in 20-60% of patients, histological transformation to DLBCL [transformed FL (tFL)] may occur, which reduces life expectancy to 1.2 years (4,6,7).

The genetic hallmark of FL is the t(14;18)(q32;q21) chromosomal aberration, identifiable in 80-90% of FL cases (8,9). This translocation juxtaposes the BCL2 gene on chromosome 18 to the enhancer sequences of the immunoglobulin heavy chain gene (IGH) promoter region on chromosome 14 (9-11), leading to the overexpression of BCL2 in B cells (12) (Fig. 1B). BCL2 normally inhibits apoptosis. BCL2 overexpression resulting from t(14;18) translocation contributes to the development of FL by preventing the apoptosis of neoplastic cells (2,13-16) However, the t(14;18) translocation can also be detected at a low level in the blood of healthy individuals (17), indicating that the translocation alone is insufficient for the development of FL. Numerous genetic alterations (Table I), many affecting tumor suppressor genes (TSGs), are additional factors in the pathogenesis of FL (Fig. 1B) (18) Chromatin modifiers, such as MLL2 and the transcription factor, interferon regulatory factor 4 (IRF4), can be mutated or deleted, affecting the molecular structure and transcription of DNA. Alterations in cell cycle regulators, such as CDKN2A, which activates the p53 and pRb pathways, can also occur. Signal transduction processes can also be affected by mutations in genes such as FAS, which also plays a role in apoptosis (18).

Table I

Genes altered in follicular lymphoma.

Table I

Genes altered in follicular lymphoma.

GenePathway affected(Refs.)
ARID1ATranscription regulation; member of the SWI-SNF family(75,76)
BCL2Apoptosis regulator protein(11)
BCL6Apoptosis regulator protein(77)
CDKN2AActivation of p53 and pRb pathways(75,78-84)
CREBBPActivation of transcription(85,86)
DAPK1Gamma-interferon induced apoptosis(87)
EBF1Transcription factor(75,86)
EP300Regulation of transcription by chromatin remodeling(82,88)
EPHA7Protein tyrosine kinase family(81)
EZH2DNA methylation and transcriptional regulation(89)
TNFRSF6Apoptosis(82,90)
FLCNmTOR; tumor suppressor(91)
FOXO1Transcription factor(82)
HIST1H1ENuclear protein involved in nucleosome structure(76,82)
IGHV, IGLVImmunoglobulin heavy chains(92,93)
KMT2D (MLL2)Trithorax-group histone methyltransferase(85,88,89)
PAX5Transcription factor involved in neoplastic transformation(82)
PRDM1Repressor of β-interferon gene expression(75)
RB1Tumor suppression(81)
PTPN6 (SHP1)Protein tyrosine phosphatase family(94-96)
SOCS1STAT inhibition(79,82,86,97-99)
TNFAIP3Inhibition of NF-κB activation and TNF-mediated apoptosis(75,82,86)
TNFRSF14Member of TNF-receptor superfamily; component of pathways involved in activation of the immune response(82,100,101)
TP53Tumor suppression(75,82,102,103)

While the majority of FL cases are t(14;18)-positive, 10-20% of FL cases lack the translocation (19,20). Several recent studies have focused on the biology and clinical features of t(14;18)-negative FL. Apart from BCL2, the role of other transcription factors or regulators, such as BCL6 expression, has been investigated (21,22). This review summarizes the current knowledge of the molecular characteristics of t(14;18)-negative FL. The review also describes the diagnosis and grading of t(14;18)-negative FL and summarizes the clinical features of the various subtypes.

2. Molecular characterization of t(14;18)-negative FL

Grading of FL

Cases of FL are generally classified into 1 of 3 grades based on the guidelines of the World Health Organization (WHO) (23). The grading of FL is based on the average number of centroblasts in 10 randomly selected microscopic x40 high power fields (HPFs). In grade 1 FL, up to 5 centroblasts per HPF are present; grade 2 FL has 6 to 15 centroblasts per HPF, and grade 3 FL has >15 centroblasts per HPF. Grade 3 is further subdivided into 3A, in which some centrocytes are present, and 3B with no centrocytes (24). Approximately 25% of FL patients are in grade 1, 25-50% in grade 2, and ≥50% in grade 3 (25). FL grades 1 and 2 are considered low grade and affected patients do not necessarily require chemotherapy. For grade 3 FL, chemotherapy is often prescribed (24). Approximately 10% of cases of low-grade FL lack t(14;18). However, approximately 30-40 and 70-85% of grades 3A and 3B FL, respectively, are t(14;18)-negative (26,27).

Unlike grade 3A FL, grade 3B FL is less often found to be BCL2-positive by immunostaining and is associated with an increased p53 and MUM1/IRF expression (28). Grade 3B FL is positive for CD10, also known as common acute lymphocytic leukemia antigen (CALLA), in approximately 50% of cases (29), and is associated with translocations in the 3q27 locus, which affects BCL6 expression (30). In these respects, grade 3B FL more closely resembles DLBCL than the other grades of FL (3,26,27,30,31).

Presence of the t(14;18) translocation in varying grades of FL

Vaandrager et al (32) used a FISH assay to detect sequences flanking the BCL2 gene and found that the absence of t(14;18) was more common in FL grades 2 and 3. Swerdlow et al (23) also found that a lack of BCL2 overexpression was particularly common in grade 3 FL. Grade 3B FLs are composed entirely of centroblasts and are typically associated with a lack of t(14;18) (5,27,30). However, results on the frequencies of different grades in t(14;18)-negative FL have been somewhat contradictory. Perhaps owing to small sample sizes, studies, such as those by Weinberg et al (33) and Horsman et al (21) have found that t(14;18)-negative FLs were more likely to be grades 1 and 2 than grade 3.

BCL2 expression in t(14;18)-negative FL

The t(14;18) translocation leads to the overexpression of BCL2. However, in t(14;18)-negative FL, BCL2 overexpression is more variable (22,32,34-37). Leich et al (34,35) studied nodal FL grades 1, 2 and 3A, and found that t(14;18)-positive and t(14;18)-negative FL were morphologically indistinguishable. A small subset of t(14;18)-negative FL samples exhibit evidence of BCL2 overexpression, indicating that factors other than the t(14;18) translocation regulate its expression. The majority of t(14;18)-negative FL cases in this previous study (34) expressed BCL6, CD10, and IRF8. In the study by Horsman et al (21) 49 t(14;18)-negative FL cases were analyzed by immunohistochemistry. These researchers found that BCL2 was expressed in 33% of cases. In t(14;18)-negative FLs without BCL2 expression, Leich et al (35) performed microRNA expression analysis and found a decreased expression of T cell leukemia/lymphoma 1A (TCL1A), supporting the hypothesis that this subtype of FL has a late germinal center B cell phenotype.

BCL6 expression in t(14;18)-negative FL

In addition to BCL2, BCL6 is also frequently expressed in FL (37). While an absence of BCL2 overexpression is a consistent finding in t(14;18)-negative FL, BCL6 expression often accompanies the lack of BCL2 expression (38). In the study by Horsman et al (21), only 33% of 49 t(14;18)-negative FL cases expressed BCL2; however, in the majority of cases, a t(3;14) translocation was present and BCL6 was overexpressed. Jardin et al (39) examined a group of patients with t(14;18)-negative FL who had a 3q27/BCL6 gene rearrangement. Tumor cells in that study were generally negative for CD10, BCL2, CD5, CD23 and CD43, and positive for CD20 and BCL6. In that sample, follicles were significantly larger than those found in t(14;18)-positive FL. Leich et al also found that their sample of grades 1-3A t(14;18)-negative FL lacked CD10 expression (34). Jardin et al (39) speculated that an anti-apoptotic function of BCL6 may act as a surrogate for BCL2 in this group. Gollub et al (40) also found that their samples of t(14;18)-negative FLs with BCL6 translocations were more common in higher grade FL. A total of 60% of their sample of 5 t(14;18)-negative FLs with BCL6 translocations were positive for CD10. However, 100% of their sample of t(14;18)-positive FLs with BCL6 translocations also expressed CD10. In the study by Gollub et al (40), t(14;18)-negative FLs with BCL6 translocations involved the bone marrow less often and did not infiltrate the lymph node capsule as often when compared to t(14;18)-positive FLs. BCL6 translocations in that study affected the phenotype only when they were not accompanied by BCL2 translocations.

Gu et al (19) also found that BCL6 rearrangement was more common in t(14;18)-negative FL than in t(14;18)-positive FL and that BCL6 rearrangements were present in all grades of FL, but were more common in FL grade 3. Pan et al (41) also found that the BCL6 translocation was related to high-grade FL. These studies indicate that BCL6 rearrangement may play a similar role to BCL2 rearrangement in cases where that does not occur. A summary of genes mutated or abnormally expressed in t(14;18)-negative FL is shown in Table II.

Table II

Genes mutated or abnormally expressed in t(14;18)-negative follicular lymphoma.

Table II

Genes mutated or abnormally expressed in t(14;18)-negative follicular lymphoma.

GeneExpression levelSamples (%)(Refs.)
BCL2No expression64.7-100(34,35,70-72)
BCL2Overexpression20-80(34,39,45)
BCL6Expression54.5-100(34,37,39,42,70-72)
CD3Expression100(71)
CD5No expression100(39)
CD10No expression31.6-100(34,35,70)
CD10Expression26.7-100(34,39,72)
CD19Expression100(72)
CD20Expression100(70-72)
CD23Expression86.7-100(64,72)
CD43No expression100(70)
CD79aExpression100(38)
IRF8No expression2.6(34)
IRF8Expression97(34)
MUM1/IRFNo expression100(72)
TCL1ABelow normal expression67(35)
Molecular features of atypical forms of t(14;18)-negative FL

An atypical form of FL lacking the BCL2 rearrangement that differs from other examples of t(14;18)-negative FL was reported by Karube et al (42) This FL subtype exhibited an increased expression of IRF4/MUM1, a protein involved in plasma cell differentiation. The samples were also negative for CD10. Patients with CD10-MUM1+ FL were likely to be more elderly than typical FL patients, and more often had grade 3A or 3B morphology and diffuse proliferation. Additionally, 88% of these FLs exhibited BCL6 translocation or amplification.

In a study on the molecular features of t(14;18)-negative FL, Gagyi et al (43) found that t(14;18)-negative FL exhibited no difference when compared to t(14;18)-positive FL in the presence of somatic hypermutations of IGHV genes and the levels of expression of activation-induced cytidine deaminase (AID). This supported a germinal center origin and differentiation in both groups of FL. Additionally, there were no differences in somatic hypermutations of the MYC, RhoH, or PAX5 genes between the 2 groups. These results led Gagyi et al to the conclusion that t(14;18)-positive and t(14;18)-negative should be considered as a single entity. The authors state that, in some cases of t(14;18)-negative FL, BCL2 can be overexpressed by genetic events other than translocation, and other cases of t(14;18)-negative FL, rearrangement of the BCL6 gene could provide the basis of substitution for the lack of BCL2 expression (43).

Another distinct form of t(14;18)-negative FL is pediatric-type FL (44-47). This form of FL is characterized by highly proliferative follicles, which often have blastoid follicular center cells instead of classic centroblasts (44). These cases are negative for BCL2, BCL6, and MYC rearrangements (45). In this form of FL, dissemination beyond the skin is uncommon, and the cases usually have a good clinical prognosis (46). Although previously associated with a primarily pediatric population, there are occurrences in adults (44,47). The study by Martin-Guerrero et al (45) demonstrated that mutations of tumor necrosis factor receptor superfamily 14 (TNFRSF14) accompanied by copy-number neutral loss of heterozygosity (CNN-LOH) at the 1p36 site were more common in pediatric-type FL than in a comparison group with non-pediatric t(14;18)-negative FL. Thus, genetic alterations at the 1p36 site may underlie pathogenesis in pediatric-type FL. The variations in symptomology and gene rearrangements within the t(14;18)-negative FL patient population underscores the diversity of the disease and difficulties related to a coherent classification. These issues suggest that better diagnostic methods are warranted.

3. Genetic abnormalities recently identified through next-generation sequencing

Through the use of next-generation sequencing techniques in recent years, researchers have concluded that epigenetic mechanisms of gene regulation play an important role in the development of FL (48). Among the most important and consistent results from these next-generation sequencing studies has been the identification of an important role for histone methyltransferases KMT2D and EZH2 in epigenetic mutations (Table I) (48,49). Next-generation sequencing has also been used to identify the important role of TNFRSF14, which is abnormally expressed in about 40% of FL patients (50). TNFRSF14 encodes a gene termed herpes virus entry mediator, which acts to limit T-cell activation (Table I) (50).

4. Treatment and clinical outcomes

Clinical staging of lymphomas

Various treatment regimens exist for FL, and the appropriate treatment strategy depends upon the grade and clinical characteristics of the disease. A clinical staging system for lymphomas was published by the Committee on Hodgkin's Disease Staging Classification (51). In this system, stage I indicates the involvement of a single lymph node region or a single extranodal site in the cancer. Stage II indicates the involvement of two or more separate lymph node regions on the same side of the diaphragm. Stage III indicates the involvement of lymph node regions on both sides of the diaphragm. Stage IV indicates diffuse or disseminated involvement of one or more extralymphatic organs. Stage IV also includes localized involvement of liver, bone marrow, or nodular involvement of the lungs.

Approximately 26-33% of FL patients are in stage I or II (52). Approximately 20% of patients with FL undergo spontaneous remission without any treatment (53). In approximately 30-40% of patients with FL, a histological transformation to DLBCL occurs, and the majority patients with histological transformation succumb to the disease within a year (54). In terms of histological transformation and the overall survival of patients, there are no reliably documented differences between patients with t(14;18)-positive and t(14;18)-negative FL (55). As no reliable correlation between BCL2 gene rearrangement status and clinical outcome has been reported, the t(14;18) translocation has not been used as a prognostic marker (56). Furthermore, the results of numerous studies have suggested that a similar set of morphologic, immunophenotypic, and clinical characteristics develop in FL regardless of the presence or absence of a BCL2 rearrangement in FL grades 1-3A. Thus, the generally accepted opinion of these grades of FL is that they constitute a homogenous group with some distinct but many common molecular pathways (43). A depiction of the major targets of drug treatments in t(14;18)-positive and t(14;18)-negative FL is illustrated in Fig. 2. Furthermore, a summary of patient groups with t(14;18)-negative FL with their treatments and outcomes is illustrated in Table III.

Table III

Groups of patients with t(14;18)-negative FL with treatments and outcomes.

Table III

Groups of patients with t(14;18)-negative FL with treatments and outcomes.

Patient groupTreatmentOutcome(Refs.)
Five-hundred and forty patients with advanced-stage FL and 116 patients with early-stage FL. Some patients with BCL2 translocations and others without BCL2 translocations.Chemotherapeutic regimens and radiation.Patient characteristics did not differ between patients with and without BCL2 translocations. Survival times of patients with advanced-stage FL did not differ.(66)
Seventy-seven patients with FL and 22 patients with DLBCL with BCL2 translocations. Fourteen patients with FL and 14 patients with DLBCL with BCL6 translocations.R-CHOPPatients were evaluated at 29 months following treatment. Overall survival and progression-free survival did not differ between groups.(68)
Fifteen patients with t(14;18)-negative FL with BCL6 translocations. The majority of patients in the group had a t(3;14) translocation.CHOP, radiotherapy, or chlorambucilWhen compared to t(14;18)-positive controls, there was no difference in age, sex, performance status, bone marrow involvement, or overall survival.(39)
Thirty-five cases of predominantly diffuse t(14;18)-negative FLRadiotherapy, chemotherapy in combination with radiotherapy, or chemotherapy alone.Therapy data were available for 19 patients. Fifteen had complete remission and one had progressive disease.(5)
Fifty-one-year-old male with t(14;18)-negative FL who lacked BCL2 expression.Examination of lymph node biopsy.Diagnosis of high-grade 3A FL with DLBCL transformation.(70)
Seven patients, including 5 females and 2 males. The cases had centroblast-predominant isolated follicles and a lack of BCL2 staining in otherwise-normal lymph nodes.Clinical examination, including lymph node samples, bone marrow biopsies, imaging studies, and clinical evaluations.None of the patients exhibited evidence of systematic lymphoma at the time of diagnosis or clinical follow-up visits up to 44 months after diagnosis.(71)
One patient with t(14;18)-negative FL. The patient was a 74-year-old female with swelling in the submandibular region.A surgery was performed and the lymph node was removed.No recurrence of FL upon follow-up one year after surgery.(72)
Common treatment strategies

Unless the disease can be cured by surgery or local irradiation, FL is generally managed with radiation or chemotherapy. For those projected for treatment with radiation or chemotherapy, clinical monitoring using a 'watch and wait' approach is likely to be recommended until the patient requires treatment (1). For patients who have stage I or II low-grade FL, radiation is usually recommended as a first treatment approach. For patients with low-grade FL with a high tumor burden, chemotherapy is often followed by involved field irradiation therapy (IFRT) (22). In stage III or IV FL, patients typically receive chemotherapy regimens, such as alkylating agents for inhibition of cell growth, purine analogs and anthracycline-based agents (22). Monochemotherapy with chlorambucil or cyclophosphamide is a common approach (22). Treatment with chemotherapy plus rituximab is the accepted standard for most patients and this method is associated with increases in the overall survival of patients (2). Among anthracycline-based regimens, one of the most common treatments is R-CHOP, a combination of 5 drugs: Rituximab, cyclophosphamide (Cytoxan), doxorubicin (Adriamycin), vincristine and prednisone (57). Another commonly used combination chemotherapy is R-CVP, which includes rituximab, cyclophosphamide, vincristine and prednisone (58). CHOP-RIT, the administration of tositumomab/iodine-131 radioimmunotherapy (targets CD20) immediately following CHOP, is also common (59). Another commonly used approach is bendamustine (an alkylating agent) plus rituximab (60). However, rituximab, a monoclonal anti-CD20 antibody, is ineffective in variants of FL that are negative for CD20 (61,62). A recently developed approach is ABT199, a BH3 mimetic investigational drug (venetoclax) that targets the BCL2 pathway (63). However, as the majority of cases of t(14;18)-negative FL lack BCL2 overexpression, this approach would be contraindicated for these patients. A better understanding of the underlying genetics of each variation on the disease may lead to more precisely targeted treatment regimens.

Major signal transduction pathways targeted in drug treatment for FL

Although the exact mechanisms of rituximab, the most common drug treatment for FL, are not yet fully known (64), it is considered that rituximab achieves its clinical effect by altering intracellular signaling pathways (64). Evidence indicates that rituximab operates by reducing activity of the p38MAPK pathway. This reduction in activity results in the inhibition of the interleukin-10 paracrine cytokine auto-regulatory loop, which in turn inhibits signal transducer and activator of transcription 3 (STAT-3) activity. This inhibition of STAT-3 activity leads to a downregulation of BCL2 expression, and subsequent chemosensitization (65). Additionally, rituximab upregulates expression of Raf-1 kinase inhibitor protein (RKIP) (64). RKIP acts to reduce the activity of the extracellular signal-related kinase (ERK) 1/2 and the NF-κB pathways, which in turn inhibits transcription of AP-1 and NF-κB. The downregulation of both of these pathways leads to the subsequent reduction of BCL2 transcription and expression and susceptibility to apoptosis (64). A better understanding of the effects of drugs on molecular pathways will allow better targeted treatments.

Clinical differences between t(14;18)-positive and -negative FL

The research group of Leich et al (34,35,66) conducted studies examining clinical differences between patients with t(14;18)-negative and t(14;18)-positive FL. In one study comparing the 2 groups, t(14;18)-negative patients were more likely to have lower stage FL. However, no differences were observed in the other clinical variables of sex, involvement of extranodal sites, B symptoms, lactate dehydrogenase and the Eastern Cooperative Oncology Group (ECOG) performance status (34,67). In another study examining 540 advanced stage and 116 early stage patients with FL treated with chemotherapy and radiation, no differences were observed in patient characteristics between FL cases with and without t(14;18). The survival times patients of patients with advanced-stage FL also did not differ between the t(14;18)-positive and t(14;18)-negative patients (66). Larger population studies should better inform treatment strategies by identifying subtle differences between the t(14;18)-negative and t(14;18)-positive FL populations.

Relation between BCL2 and BCL6 expression and clinical variables

Studies have examined the clinical differences between patients with and without BCL2 and BCL6 rearrangements. Although the majority of results underscore a similarity in clinical presentation between patients with and without BCL2 and BCL6 rearrangements, some differences have been noted. A total of 98 patients with FL and 93 DLBCL were examined by in the study by Watanabe et al (68) with the aim of determining the frequencies of BCL6 and BCL2 translocations in the two patient groups. BCL2 translocations were detected in 77 patients with FL and 22 patients with DLBCL. BCL6 translocations were detected in 14 patients with FL and 14 patients with DLBCL. The patients were followed-up for 29 months after undergoing R-CHOP therapy. For patients with FL and DLBCL, the presence of BCL2 or BCL6 translocation was uncorrelated with clinical outcomes after treatment with R-CHOP therapy. Furthermore, neither overall survival nor progression free survival differed between the patient groups (68).

Some clinical differences between t(14;18)-negative FL patients with BCL6 rearrangements and t(14;18)-positive FL patients have been noted (39). In one study, Jardin et al (39) examined a group of 15 t(14;18)-negative FL patients with BCL6 gene rearrangements and a t(14;18)-positive comparison group. A majority of the t(14;18)-negative group had a t(3;14) translocation. There were no significant differences in age, sex, performance status, bone marrow involvement, or overall survival in comparison to a t(14;18)-positive group. However, in the t(14;18)-negative group, patients were more likely to have a stage III or IV disease. Furthermore, the lymphomas in the t(14;18)-negative group had a prominent nodal architecture, and usually had significantly larger follicles than the t(14;18)-positive FLs. The authors (39) concluded from this that BCL6 may play a role in follicular structure. These results indicated that t(14;18)-negative FL with a BCL6 translocation may constitute a distinct subtype of FL.

t(14;18)-negative FL with diffuse and skin-localized expression

Studies have identified uncommon forms of t(14;18)-negative FL, some with diffuse, extranodal expression, and others with expression limited to the skin. One study demonstrated that a subtype of CD10MUM1+ FL exhibited distinctive biological and clinical features compared with the more typical CD10+MUM1 FL (42). Twenty-two mostly t(14;18)-negative FL patients who were negative for expression of CD10 and positive for expression of MUM1 were generally older than typical FL patients (mean of 67 vs. 58.7 years old). CD10-MUM1+ FL patients also exhibited diffuse FL proliferation and were generally of a higher disease grade, either FL 3A or 3B, having a relatively poor prognosis.

In another study, a different subtype of t(14;18)-negative nodal follicular non-Hodgkin lymphoma exhibited a predominantly diffuse pattern and was characterized by deletions in the chromosomal region 1p36. These patients were more frequently diagnosed as low-grade (stage I and II) FL and generally presented with isolated large tumors (median size of 5 cm) that were frequently localized to the inguinal region. Although a complete remission could be achieved by treatment with radiotherapy alone or chemotherapy in combination with radiotherapy, disease progression generally followed an indolent course (5). This diffuse form of t(14;18)-negative FL represents a previously undocumented form of FL with distinct molecular and clinical features (5).

Moreover, a subtype of generally t(14;18)-negative FL which arises primarily in the skin has also been documented (69). Each of the evaluated 47 patients with FL was in clinical stage I. Compared with t(14;18)-positive cases, these FL cases were less likely to express BCL2 or CD10. These cases also had a higher proportion of extranodal FL sites, and had a significantly higher overall and disease specific five-year survival rate. This subtype of FL has a generally more favorable clinical outcome when compared to t(14;18)-positive FL (19,69).

Clinical reports of atypical t(14;18)-negative FL

An unusual case, difficult to diagnose, was reported by Wong et al (70). The authors reported a case of a 51-year-old male with t(14;18)-negative FL who lacked BCL2 expression. The patient was classified as having high-grade 3A FL with areas of large cell transformation within the same lymph node. Neoplastic cells from the patient were positive for CD20, CD79a and BCL6, and were negative for BCL2 and CD10. The clinical diagnosis was FL 3A with diffuse large B cell transformation.

Another report of atypical clinical cases with difficult diagnoses was made by Nybakken et al (71). These researchers reported on 7 patients (5 females ranging in age from 51 to 61 years, and 2 males aged 6 and 61 years). The patients presented with isolated atypical follicles that were composed of cytologically atypical centroblasts located within otherwise normal lymph nodes. The isolated follicles had unusual morphologic features that could be mistaken for lymphoma. The cases were generally positive for CD20 and BCL6, and were negative for BCL2 and CD3. The sites of involvement included peripheral and internal sites, such as a periaortic lymph node and rectal mucosa-associated lymphoid tissue. The authors considered a diagnosis of partial involvement by FL. The cytological characteristics of involved areas were consistent with FL grade 3B (enlarged with vesicular chromatin and prominent nucleoli, and nuclear membrane irregularity inconsistent with the morphology of the surrounding lymph node). However, the pattern of single scattered follicles was not consistent with partial FL. Furthermore, none of the patients had evidence of systematic lymphoma at the time of diagnosis or clinical follow-up visits up to 44 months after diagnosis, and the patients did not require aggressive clinical management. Nybakken et al (71) noted that it is of high importance to differentiate these cases from aggressive FL or a similar disease to avoid unnecessarily aggressive treatment of patients with indolent conditions.

Surgical treatment for t(14;18)-negative FL

Finally, a surgical treatment for a patient with t(14;18)-negative FL has also been reported (72). The patient was a 74-year-old Japanese female who presented with a complaint of swelling in the submandibular region. The patient was diagnosed with stage I FL. A surgery was performed and the lymph node was removed successfully. Flow cytometric analysis revealed that the lymphoma cells were positive for CD19, CD20, CD23, CD10 and BCL6. The cells were negative for BCL2 and MUM1. There was no recurrence of FL upon follow-up a year post-surgery. This case suggests a distinct type of surgical treatment for certain types of BCL2-negative FL (72).

5. Conclusions

The t(14;18) translocation is absent in approximately 10-20% of FL, a condition less common in FL grades 1 and 2 and more common in FL grades 3A and 3B. t(14;18)-negative FL is often associated with a t(3;14) translocation or the overexpression of BCL6, which is associated with morphological effects that closely resemble those associated with t(14;18). t(14;18)-negative FL patients are generally indistinguishable from t(14;18)-positive patients from a clinical perspective. However, diffuse and pediatric forms of FL, which often lack t(14;18), have a better clinical prognosis than t(14;18)-positive FL patients. In the future, the development of novel methods for targeting pathways other than the BCL2 pathway to effectively manage t(14;18)-negative FL may prove to be useful. For example, the recent study by Cardenas et al (73) detailed the development of specific BCL6 inhibitors, such as FX1, which could be effective in controlling t(14;18)-negative FL (Fig. 2).

Acknowledgments

Not applicable.

Funding

This study was supported in part by grants from Colleges and Universities Key Scientific Research Project Plan Basic Research Special of Henan Province (grant no. 19zx009), Science and Technology Project for Tackling Key Problems of Henan Province (grant no. 182102311172), Basic and Frontier Technology Research Program of Henan Province (grant no. 132300410448), and Science and Technology Innovation Talent Program of Henan Province (grant no. 154100510019).

Availability of data and materials

Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.

Authors' contributions

WR, ST, DZ and PL contributed to the conception and design of this study. WR, ST, ZuZ, TL, XZ and ZhZ were involved in the acquisition of the data and the study design. WR, ST, ZuZ, TL, XZ, ZhZ, DZ and PL were involved in the writing of the article. WR, ST critically revised the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Follicular Lymphoma, Pamphlet. Lymphoma Association; 2017

2 

Abd El-Ghaffar HA, Shamaa S, Attwan N, et al: Stratification of Patients with Follicular Lymphoma. IntechOpen; Rijeka: 2012, https://www.intechopen.com/books/hematology-science-and-practice/stratification-of-patients-with-follicular-lymphoma. Accessed March 2, 2012.

3 

Kridel R, Sehn LH and Gascoyne RD: Pathogenesis of follicular lymphoma. J Clin Invest. 122:3424–3431. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Bisikirska B, Bansal M, Shen Y, Teruya-Feldstein J, Chaganti R and Califano A: Elucidation and pharmacological targeting of novel molecular drivers of follicular lymphoma progression. Cancer Res. 76:664–674. 2016. View Article : Google Scholar :

5 

Katzenberger T, Kalla J, Leich E, Stöcklein H, Hartmann E, Barnickel S, Wessendorf S, Ott MM, Müller-Hermelink HK, Rosenwald A and Ott G: A distinctive subtype of t(14;18)-negative nodal follicular non-Hodgkin lymphoma characterized by a predominantly diffuse growth pattern and deletions in the chromosomal region 1p-36. Blood. 113:1053–1061. 2009. View Article : Google Scholar

6 

Casulo C, Burack WR and Friedberg JW: Transformed follicular non-Hodgkin lymphoma. Blood. 125:40–47. 2015. View Article : Google Scholar

7 

Christie L, Kernohan N, Levison D, Sales M, Cunningham J, Gillespie K, Batstone P, Meiklejohn D and Goodlad J: C-MYC translocation in t(14;18) positive follicular lymphoma at presentation: An adverse prognostic indicator? Leuk Lymphoma. 49:470–476. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Bakhski A, Jensen JP, Goldman P, Wright JJ, McBride OW, Epstein AL and Korsmeyer SJ: Cloning the chromosomal break-point of t(14;18) human lymphomas: Clustering around Jh on chromosome 14 and near a transcriptional unit on 18. Cell. 41:899–906. 1985. View Article : Google Scholar

9 

Tsujimoto Y, Gorham J, Cossman J, Jaffe E and Croce CM: The t(14;18) chromosome translocations involved in B-cell neoplasms result from mistakes in VDJ joining. Science. 229:1390–1393. 1985. View Article : Google Scholar : PubMed/NCBI

10 

Cleary ML and Sklar J: Nucleotide sequence of a t(14;18) chromosomal breakpoint in follicular lymphoma and demonstration of a breakpoint-cluster region near a transcriptionally active locus on chromosome 18. Proc Natl Acad Sci USA. 82:7439–7443. 1985. View Article : Google Scholar : PubMed/NCBI

11 

Tsujimoto Y, Finger LR, Yunis J, Nowell PC and Croce CM: Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation. Science. 226:1097–1099. 1984. View Article : Google Scholar : PubMed/NCBI

12 

Graninger WB, Seto M, Boutain B, Goldman P and Korsmeyer SJ: Expression of Bcl-2 and Bcl-2-Ig fusion transcripts in normal and neoplastic cells. J Clin Invest. 80:1512–1515. 1987. View Article : Google Scholar : PubMed/NCBI

13 

Godon A, Moreau A, Talmant P, Baranger-Papot L, Geneviéve F, Milpied N, Zandecki M and Avet-Loiseau H: Is t(14;18)(q32;q21) a constant finding in follicular lymphoma? An interphase FISH study on 63 patients Leukemia. 17:255–259. 2003.

14 

Husson H, Carideo EG, Neuberg D, Schultze J, Munoz O, Marks PW, Donovan JW, Chillemi AC, O'Connell P and Freedman AS: Gene expression profiling of follicular lymphoma and normal germinal center B cells using cDNA arrays. Blood. 99:282–289. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Masir N, Campbell LJ, Goff LK, Jones M, Marafioti T, Cordell J, Clear AJ, Lister TA, Mason DY and Lee AM: BCL2 protein expression in follicular lymphomas with t(14;18) chromosomal translocations. Br J Haematol. 144:716–725. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Staudt LM: A closer look at follicular lymphoma. N Engl J Med. 356:741–742. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Roulland S, Kelly RS, Morgado E, Sungalee S, Solal-Celigny P, Colombat P, Jouve N, Palli D, Pala V, Tumino R, et al: t(14;18) Translocation: A predictive blood biomarker for follicular lymphoma. J Clin Oncol. 32:1347–1355. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Graham C and LeBrun DP: Tumor suppressors in follicular lymphoma. Leuk Lymphoma. 56:1981–1988. 2015. View Article : Google Scholar

19 

Gu K, Fu K, Jain S, Liu Z, Iqbal J, Li M, Sanger WG, Weisenburger DD, Greiner TC, Aoun P, et al: t(14;18)-negative follicular lymphomas are associated with a high frequency of BCL6 rearrangement at the alternative breakpoint region. Mod Pathol. 22:1251–1257. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Küppers R and Dalla-Favera R: Mechanisms of chromosomal translocations in B cell lymphomas. Oncogene. 20:5580–5594. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Horsman DE, Okamoto I, Ludkovski O, Le N, Harder L, Gesk S, Siebert R, Chhanabhai M, Sehn L, Connors JM and Gascoyne RD: Follicular lymphoma lacking the t(14;18)(q32;q21): Identification of two disease subtypes. Br J Haematol. 120:424–433. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Vitolo U, Ferreri AJ and Montoto S: Follicular lymphomas. Crit Rev Oncol Hematol. 66:248–261. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, Advani R, Ghielmini M, Salles GA, Zelenetz AD and Jaffe ES: The 2016 revision of theWorld Health Organization classification of lymphoid neoplasms. Blood. 127:2375–2390. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Samsi S, Lozanski G, Shana'ah A, Krishanmurthy AK and Gurcan MN: Detection of follicles from IHC-stained slides of follicular lymphoma using iterative watershed. IEEE Trans Biomed Eng. 57:2609–2612. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Paik PK, Johnson ML, D'Angelo SP, Sima CS, Ang D, Dogan S, Miller VA, Ladanyi M, Kris MG and Riely GJ: Driver mutations determine survival in smokers and never-smokers with stage IIIB/IV lung adenocarcinomas. Cancer. 118:5840–5847. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Katzenberger T, Ott G, Klein T, Kalla J, Müller-Hermelink HK and Ott MM: Cytogenetic alterations affecting BCL6 are predominantly found in follicular lymphomas grade 3B with a diffuse large B-cell component. Am J Pathol. 165:481–490. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Ott G, Katzenberger T, Lohr A, Kindelberger S, Rüdiger T, Wilhelm M, Kalla J, Rosenwald A, Müller JG, Ott MM and Müller-Hermelink HK: Cytomorphologic, immunohistochemical, and cytogenetic profiles of follicular lymphoma: 2 types of follicular lymphoma grade 3. Blood. 99:3806–3812. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Horn H, Schmelter C, Leich E, Salaverria I, Katzenberger T, Ott MM, Kalla J, Romero M, Siebert R, Rosenwald A and Ott G: Follicular lymphoma grade 3B is a distinct neoplasm according to cytogenetic and immunohistochemical profiles. Haematologica. 96:1327–1334. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Salaverria I and Siebert R: Follicular lymphoma grade 3B. Best Pract Res Clin Haematol. 24:111–119. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Bosga-Bouwer AG, van Imhoff GW, Boonstra R, van der Veen A, Haralambieva E, van den Berg A, Jong B, Krause V, Palmer MC, Coupland R, et al: Follicular lymphoma grade 3B includes 3 cytogenetically defined subgroups with primary t(14;18), 3q27, or other translocations: t(14;18) and 3q27 are mutually exclusive. Blood. 101:1149–1154. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Shustik J, Han G, Farinha P, Johnson NA, Ben Neriah S, Connors JM, Sehn LH, Horsman DE, Gascoyne RD and Steidl C: Correlations between BCL6 rearrangement and outcome in patients with diffuse large B-cell lymphoma treated with CHOP or R-CHOP. Haematologica. 95:96–101. 2010. View Article : Google Scholar :

32 

Vaandrager JW, Schuuring E, Raap T, Philippo K, Kleiverda K and Kluin P: Interphase FISH detection of BCL2 rearrangement in follicular lymphoma using breakpoint-flanking probes. Genes Chromosomes Cancer. 27:85–94. 2000. View Article : Google Scholar

33 

Weinberg OK, Ai WZ, Mariappan MR, Shum C, Levy R and Arber DA: 'Minor' BCL2 breakpoints in follicular lymphoma: Frequency and correlation with grade and disease presentation in 236 cases. J Mol Diagn. 9:530–537. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Leich E, Salaverria I, Bea S, Zettl A, Wright G, Moreno V, Gascoyne RD, Chan WC, Braziel RM, Rimsza LM, et al: Follicular lymphomas with and without translocation t(14;18) differ in gene expression profiles and genetic alterations. Blood. 114:826–834. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Leich E, Zamo A, Horn H, Haralambieva E, Puppe B, Gascoyne RD, Chan WC, Braziel RM, Rimsza LM, Weisenburger DD, et al: MicroRNA profiles of t(14;18)-negative follicular lymphoma support a late germinal center B-cell phenotype. Blood. 118:5550–5558. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Schraders M, de Jong D, Kluin P, Groenen P and van Krieken H: Lack of Bcl-2 expression in follicular lymphoma may be caused by mutations in the BCL2 gene or by absence of the t(14;18) translocation. J Pathol. 205:329–335. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Skinnider BF, Horsman DE, Dupuis B and Gascoyne RD: Bcl-6 and Bcl-2 protein expression in diffuse large B-cell lymphoma and follicular lymphoma: Correlation with 3q27 and 18q21 chromosomal abnormalities. Hum Pathol. 30:803–808. 1999. View Article : Google Scholar : PubMed/NCBI

38 

Guo Y, Karube K, Kawano R, Suzumiya J, Takeshita M, Kikuchi M, Huang GS, Li Q and Ohshima K: Bcl2-negative follicular lymphomas frequently have Bcl6 translocation and/or Bcl6 or p53 expression. Pathol Int. 57:148–152. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Jardin F, Gaulard P, Buchonnet G, Contentin N, Lepretre S, Lenain P, Stamatoullas A, Picquenot JM, Duval C, Parmentier F, et al: Follicular lymphoma without t(14;18) and with BCL-6 rearrangement: A lymphoma subtype with distinct pathological, molecular and clinical characteristics. Leukemia. 16:2309–2317. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Gollub W, Stassek B, Huckhagel T, Bernd HW, Krokowski M, Merz H, Feller AC and Thorns C: BCL6-translocations affect the phenotype of follicular lymphomas only in the absence of t(14;18)IgH/BCL2. Anticancer Res. 29:4649–4655. 2009.PubMed/NCBI

41 

Pan Y, Meng B, Sun B, Guan B, Liang Y, Wang H, Hao X and Fu K: Frequencies of BCL2 and BCL6 translocations in representative Chinese follicular lymphoma patients: Morphologic, immunohistochemical, and FISH analyses. Diagn Mol Pathol. 21:234–240. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Karube K, Guo Y, Suzumiya J, Sugita Y, Nomura Y, Yamamoto K, Shimizu K, Yoshida S, Komatani H, Takeshita M, et al: CD10-MUM1 + follicular lymphoma lacks BCL2 gene translocation and shows characteristic biologic and clinical features. Blood. 109:3076–3079. 2007. View Article : Google Scholar

43 

Gagyi E, Balogh Z, Bödör C, Timár B, Reiniger L, Deák L, Csomor J, Csernus B, Szepesi A and Matolcsy A: Somatic hyper-mutation of IGVH genes and aberrant somatic hypermutation in follicular lymphoma without BCL-2 gene rearrangement and expression. Haematologica. 93:1822–1828. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Liu Q, Salaverria I, Pittaluga S, Jegalian AG, Xi L, Siebert R, Raffeld M, Hewitt SM and Jaffe ES: Follicular lymphomas in children and young adults: A comparison of the pediatric variant with usual follicular lymphoma. Am J Surg Pathol. 37:333–343. 2013. View Article : Google Scholar :

45 

Martin-Guerrero I, Salaverria I, Burkhardt B, Szczepanowski M, Baudis M, Bens S, de Leval L, Garcia-Orad A, Horn H, Lisfeld J, et al: Recurrent loss of heterozygosity in 1p36 associated with TNFRSF14 mutations in IRF4 translocation negative pediatric follicular lymphomas. Haematologica. 98:1237–1241. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Jaffe ES: The 2008 WHO classification of lymphomas: Implications for clinical practice and translational research. Hematol Am Soc Hematol Educ Program. 523–531. 2009. View Article : Google Scholar

47 

Louissaint A Jr, Ackerman AM, Dias-Santagata D, Ferry JA, Hochberg EP, Huang MS, Iafrate AJ, Lara DO, Pinkus GS, Salaverria I, et al: Pediatric-type nodal follicular lymphoma: An indolent clonal proliferation in children and adults with high proliferation index and no BCL2 rearrangement. Blood. 120:2395–2404. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Araf S, Okosun J, Koniali L, Fitzgibbon J and Heward J: Epigenetic dysregulation in follicular lymphoma. Epigenomics. 8:77–84. 2016. View Article : Google Scholar :

49 

Braggio E, Egan JB, Fonseca R and Stewart AK: Lessons from next-generation sequencing analysis in hematological malignancies. Blood Cancer J. 3:e1272013. View Article : Google Scholar : PubMed/NCBI

50 

Kotsiou E, Okosun J, Besley C, Iqbal S, Matthews J, Fitzgibbon J, Gribben JG and Davies JK: TNFRSF14 aberrations in follicular lymphoma increase clinically significant allogeneic T-cell responses. Blood. 128:72–81. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Carbone PP, Kaplan HS, Musshoff K, Smithers DW and Tubiana M: Report of the committee on Hodgkin's disease staging classification. Cancer Res. 31:1860–1861. 1971.PubMed/NCBI

52 

Armitage JO and Weisenburger DD: New approach to classifying non-Hodgkin's lymphomas: Clinical features of the major histo-logic subtypes. Non-Hodgkin's lymphoma classification project. J Clin Oncol. 16:2780–2795. 1998. View Article : Google Scholar : PubMed/NCBI

53 

Horning SJ and Rosenberg SA: The natural history of initially untreated low-grade non-Hodgkin's lymphomas. N Engl J Med. 311:1471–1475. 1984. View Article : Google Scholar : PubMed/NCBI

54 

Takata K, Miyata-Takata T, Sato Y and Yoshino T: Pathology of follicular lymphoma. J Clin Exp Hematop. 54:3–9. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Nathwani BN: A promising new biologic prognostic model in diffuse large B-cell lymphoma. Blood. 120:2161–2162. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Pezella F, Jones M, Ralfkiaer E, Ersbøll J, Gatter KC and Mason DY: Evaluation of bcl-2 protein expression and 14;18 translocation as prognostic markers in follicular lymphoma. Br J Cancer. 65:87–89. 1992. View Article : Google Scholar

57 

Tomita N, Takasaki H, Miyashita K, Fujisawa S, Ogusa E, Matsuura S, Kishimoto K, Numata A, Fujita A, Ohshima R, et al: R-CHOP therapy alone in limited stage diffuse large B-cell lymphoma. Br J Haematol. 161:383–388. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Siddhartha G and Vijay P: R-CHOP versus R-CVP in the treatment of follicular lymphoma: A meta-analysis and critical appraisal of current literature. J Hematol Oncol. 2:142009. View Article : Google Scholar : PubMed/NCBI

59 

Press OW, Unger JM, Rimsza LM, Friedberg JW, LeBlanc M, Czuczman MS, Kaminski M, Braziel RM, Spier C, Gopal AK, et al: A comparative analysis of prognostic factor models for follicular lymphoma based on a phase III trial of CHOP-rituximab versus CHOP + 131iodine-tositumomab. Clin Cancer Res. 19:6624–6632. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Kahl BS and Yang DT: Follicular lymphoma: Evolving therapeutic strategies. Blood. 127:2055–2063. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Maloney DG, Grillo-López AJ, White CA, Bodkin D, Schilder RJ, Neidhart JA, Janakiraman N, Foon KA, Liles TM, Dallaire BK, et al: IDEC-C2B8 (Rituximab) anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin's lymphoma. Blood. 90:2188–2195. 1997. View Article : Google Scholar : PubMed/NCBI

62 

McLaughlin P, Grillo-López AJ, Link BK, Levy R, Czuczman MS, Williams ME, Heyman MR, Bence-Bruckler I, White CA, Cabanillas F, et al: Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: Half of patients respond to a four-dose treatment program. J Clin Oncol. 16:2825–2833. 1998. View Article : Google Scholar : PubMed/NCBI

63 

Cang S, Iragavarapu C, Savooji J, Song Y and Liu D: ABT-199 (venetoclax) and BCL-2 inhibitors in clinical development. J Hematol Oncol. 8:1292015. View Article : Google Scholar : PubMed/NCBI

64 

Jazirehi AR and Bonavida B: Cellular and molecular signal transduction pathways modulated by rituximab (rituxan, anti-CD20 mAb) in non-Hodgkin's lymphoma: Implications in chemosensitization and therapeutic intervention. Oncogene. 24:2121–2143. 2005. View Article : Google Scholar : PubMed/NCBI

65 

Alas S, Emmanouilides C and Bonavida B: Inhibition of interleukin 10 by rituximab results in down-regulation of bcl-2 and sensitization of B-cell non-Hodgkin's lymphoma to apoptosis. Clin Cancer Res. 7:709–723. 2001.PubMed/NCBI

66 

Leich E, Hoster E, Wartenberg M, Unterhalt M, Siebert R, Koch K, Klapper W, Engelhard M, Puppe B, Horn H, et al: Similar clinical features in follicular lymphomas with and without breaks in the BCL2 locus. Leukemia. 30:854–860. 2016. View Article : Google Scholar

67 

Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET and Carbone PP: Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 5:649–655. 1982. View Article : Google Scholar : PubMed/NCBI

68 

Watanabe R, Tomita N, Matsumoto C, Hattori Y, Matsuura S, Takasaki H, Hashimoto C, Fujita H, Fujisawa S and Ishigatsubo Y: The 3q27 and 18q21 translocations for follicular lymphoma and diffuse large B-cell lymphoma in the rituximab era. J Clin Exp Hematop. 53:107–114. 2013. View Article : Google Scholar : PubMed/NCBI

69 

Goodlad JR, Batstone PJ, Hamilton DA, Kernohan NM, Levison DA and White JM: BCL2 gene abnormalities define distinct clinical subsets of follicular lymphoma. Histopathology. 49:229–241. 2006. View Article : Google Scholar : PubMed/NCBI

70 

Wong YP, Faridah AR, Ahmad Toha S and Noraidah M: A case of t(14;18)-negative follicular lymphoma with unusual immunopheno-type: A diagnostic dilemma. Med Health. 6(1 Suppl): S2072011.

71 

Nybakken GE, Bala R, Gratzinger D, Jones CD, Zehnder JL, Bangs CD, Cherry A, Warnke RA and Natkunam Y: Isolated follicles enriched for centroblasts and lacking t(14;18)/BCL2 in lymphoid tissue: Diagnostic and clinical implications. PLoS One. 11:e01517352016. View Article : Google Scholar : PubMed/NCBI

72 

Kamatani T, Mishima K, Kutsuna T, Yoshihama Y, Kondo S and Shintani S: A case of surgical treatment for primary BCL2-'negative' follicular lymphoma without t(14;18) in early stage of the submandibular lymph node. J Oral Maxillofac Surg Med Pathol. 26:599–602. 2014. View Article : Google Scholar

73 

Cardenas MG, Yu W, Beguelin W, Teater MR, Geng H, Goldstein RL, Oswald E, Hatzi K, Yang SN, Cohen J, et al: Rationally designed BCL6 inhibitors target activated B cell diffuse large B cell lymphoma. J Clin Invest. 126:3351–3362. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Ahearn IM, Haigis K, Bar-Sagi D and Philips MR: Regulating the regulator: Post-translational modification of RAS. Nat Rev Mol Cell Biol. 13:39–51. 2012. View Article : Google Scholar

75 

Bouska A, McKeithan TW, Deffenbacher KE, Lachel C, Wright GW, Iqbal J, Smith LM, Zhang W, Kucuk C, Rinaldi A, et al: Genome-wide copy-number analyses reveal genomic abnormalities involved in transformation of follicular lymphoma. Blood. 123:1681–1690. 2014. View Article : Google Scholar :

76 

Li H, Kaminski MS, Li Y, Yildiz M, Ouillette P, Jones S, Fox H, Jacobi K, Saiya-Cork K, Bixby D, et al: Mutations in linker histone genes HIST1H1 B, C, D, and E; OCT2 (POU2F2); IRF8; and ARID1A underlying the pathogenesis of follicular lymphoma. Blood. 123:1487–1498. 2014. View Article : Google Scholar : PubMed/NCBI

77 

Migliazza A, Martinotti S, Chen W, Fusco C, Ye BH, Knowles DM, Offit K, Chaganti RS and Dalla-Favera R: Frequent somatic hypermutation of the 5' noncoding region of the BCL6 gene in B-cell lymphoma. Proc Natl Acad Sci USA. 92:12520–12524. 1995. View Article : Google Scholar : PubMed/NCBI

78 

Alhejaily A, Day AG, Feilotter HE, Baetz T and Lebrun DP: Inactivation of the CDKN2A tumor-suppressor gene by deletion or methylation is common at diagnosis in follicular lymphoma and associated with poor clinical outcome. Clin Cancer Res. 20:1676–1686. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Cheung KJ, Johnson NA, Affleck JG, Severson T, Steidl C, Ben-Neriah S, Schein J, Morin RD, Moore R, Shah SP, et al: Acquired TNFRSF14 mutations in follicular lymphoma are associated with worse prognosis. Cancer Res. 70:9166–9174. 2010. View Article : Google Scholar : PubMed/NCBI

80 

Fitzgibbon J, Iqbal S, Davies A, O'Shea D, Carlotti E, Chaplin T, Matthews J, Raghavan M, Norton A, Lister TA and Young BD: Genome-wide detection of recurring sites of uniparental disomy in follicular and transformed follicular lymphoma. Leukemia. 21:1514–1520. 2007. View Article : Google Scholar : PubMed/NCBI

81 

Oricchio E, Ciriello G, Jiang M, Boice MH, Schatz JH, Heguy A, Viale A, de Stanchina E, Teruya-Feldstein J, Bouska A, et al: Frequent disruption of the RB pathway in indolent follicular lymphoma suggests a new combination therapy. J Exp Med. 211:1379–1391. 2014. View Article : Google Scholar : PubMed/NCBI

82 

Pasqualucci L, Khiabanian H, Fangazio M, Vasishtha M, Messina M, Holmes AB, Ouillette P, Trifonov V, Rossi D, Tabbò F, et al: Genetics of follicular lymphoma transformation. Cell Rep. 6:130–140. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Ross CW, Ouillette PD, Saddler CM, Shedden KA and Malek SN: Comprehensive analysis of copy number and allele status identifies multiple chromosome defects underlying follicular lymphoma pathogenesis. Clin Cancer Res. 13:4777–4785. 2007. View Article : Google Scholar : PubMed/NCBI

84 

Schwaenen C, Viardot A, Berger H, Barth TF, Bentink S, Döhner H, Enz M, Feller AC, Hansmann ML, Hummel M, et al: Microarray-based genomic profiling reveals novel genomic aberrations in follicular lymphoma which associate with patient survival and gene expression status. Genes Chromosomes Cancer. 48:39–54. 2009. View Article : Google Scholar

85 

Ogura M, Ando K, Suzuki T, Ishizawa K, Oh SY, Itoh K, Yamamoto K, Au WY, Tien HF, Matsuno Y, et al: A multicentre phase II study of vorinostat in patients with relapsed or refractory indolent B-cell non-Hodgkin lymphoma and mantle cell lymphoma. Br J Haematol. 165:768–776. 2014. View Article : Google Scholar : PubMed/NCBI

86 

Okosun J, Bödör C, Wang J, Araf S, Yang CY, Pan C, Boller S, Cittaro D, Bozek M, Iqbal S, et al: Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat Genet. 46:176–181. 2014. View Article : Google Scholar :

87 

Krajnovic M, Radojkovic M, Davidovic R, Dimitrijevic B and Krtolica K: Prognostic significance of epigenetic inactivation of p16, p15, MGMT and DAPK genes in follicular lymphoma. Med Oncol. 30:4412013. View Article : Google Scholar : PubMed/NCBI

88 

Morin RD, Mendez-Lago M, Mungall AJ, Goya R, Mungall KL, Corbett RD, Johnson NA, Severson TM, Chiu R, Field M, et al: Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature. 476:298–303. 2011. View Article : Google Scholar : PubMed/NCBI

89 

Morin RD, Johnson NA, Severson TM, Mungall AJ, An J, Goya R, Paul JE, Boyle M, Woolcock BW, Kuchenbauer F, et al: Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat Genet. 42:181–185. 2010. View Article : Google Scholar : PubMed/NCBI

90 

Grønbaek K, Straten PT, Ralfkiaer E, Ahrenkiel V, Andersen MK, Hansen NE, Zeuthen J, Hou-Jensen K and Guldberg P: Somatic Fas mutations in non-Hodgkin's lymphoma: Association with extranodal disease and autoimmunity. Blood. 92:3018–3024. 1998. View Article : Google Scholar : PubMed/NCBI

91 

Xia Q, Wang G, Wang H, Hu Q and Ying Z: Folliculin, a tumor suppressor associated with Birt-Hogg-Dube (BHD) syndrome, is a novel modifier of TDP-43 cytoplasmic translocation and aggregation. Hum Mol Genet. 25:83–96. 2016. View Article : Google Scholar

92 

Limpens J, Stad R, Vos C, de Vlaam C, de Jong D, van Ommen GJ, Schuuring E and Kluin PM: Lymphoma-associated translocation t(14;18) in blood B cells of normal individuals. Blood. 85:2528–2536. 1995. View Article : Google Scholar : PubMed/NCBI

93 

Zhu D, McCarthy H, Ottensmeier CH, Johnson P, Hamblin TJ and Stevenson FK: Acquisition of potential N-glycosylation sites in the immunoglobulin variable region by somatic mutation is a distinctive feature of follicular lymphoma. Blood. 99:2562–2568. 2002. View Article : Google Scholar : PubMed/NCBI

94 

Chim CS, Wong KY, Loong F and Srivastava G: SOCS1 and SHP1 hypermethylation in mantle cell lymphoma and follicular lymphoma: Implications for epigenetic activation of the Jak/STAT pathway. Leukemia. 18:356–358. 2004. View Article : Google Scholar

95 

Koyama M, Oka T, Ouchida M, Nakatani Y, Nishiuchi R, Yoshino T, Hayashi K, Akagi T and Seino Y: Activated proliferation of B-cell lymphomas/leukemias with the SHP1 gene silencing by aberrant CpG methylation. Lab Invest. 83:1849–1858. 2003. View Article : Google Scholar : PubMed/NCBI

96 

Oka T, Yoshino T, Hayashi K, Ohara N, Nakanishi T, Yamaai Y, Hiraki A, Sogawa CA, Kondo E, Teramoto N, et al: Reduction of hematopoietic cell-specific tyrosine phosphatase SHP-1 gene expression in natural killer cell lymphoma and various types of lymphomas/leukemias: Combination analysis with cDNA expression array and tissue microarray. Am J Pathol. 159:1495–1505. 2001. View Article : Google Scholar : PubMed/NCBI

97 

Honma K, Tsuzuki S, Nakagawa M, Tagawa H, Nakamura S, Morishima Y and Seto M: TNFAIP3/A20 functions as a novel tumor suppressor gene in several subtypes of non-Hodgkin lymphomas. Blood. 114:2467–2475. 2009. View Article : Google Scholar : PubMed/NCBI

98 

Kato M, Sanada M, Kato I, Sato Y, Takita J, Takeuchi K, Niwa A, Chen Y, Nakazaki K, Nomoto J, et al: Frequent inactivation of A20 in B-cell lymphomas. Nature. 459:712–716. 2009. View Article : Google Scholar : PubMed/NCBI

99 

Mottok A, Rennè C, Seifert M, Oppermann E, Bechstein W, Hansmann ML, Küppers R and Bräuninger A: Inactivating SOCS1 mutations are caused by aberrant somatic hypermutation and restricted to a subset of B-cell lymphoma entities. Blood. 114:4503–4506. 2009. View Article : Google Scholar : PubMed/NCBI

100 

Cheung KJ, Delaney A, Ben-Neriah S, Schein J, Lee T, Shah SP, Cheung D, Johnson NA, Mungall AJ, Telenius A, et al: High resolution analysis of follicular lymphoma genomes reveals somatic recurrent sites of copy-neutral loss of heterozygosity and copy number alterations that target single genes. Genes Chromosomes Cancer. 49:669–681. 2010. View Article : Google Scholar : PubMed/NCBI

101 

Launay E, Pangault C, Bertrand P, Jardin F, Lamy T, Tilly H, Tarte K, Bastard C and Fest T: High rate of TNFRSF14 gene alterations related to 1p36 region in de novo follicular lymphoma and impact on prognosis. Leukemia. 26:559–562. 2012. View Article : Google Scholar

102 

O'Shea D, O'Riain C, Taylor C, Waters R, Carlotti E, Macdougall F, Gribben J, Rosenwald A, Ott G, Rimsza LM, et al: The presence of TP53 mutation at diagnosis of follicular lymphoma identifies a high-risk group of patients with shortened time to disease progression and poorer overall survival. Blood. 112:3126–3129. 2008. View Article : Google Scholar : PubMed/NCBI

103 

Sander CA, Yano T, Clark HM, Harris C, Longo DL, Jaffe ES and Raffeld M: p53 mutation is associated with progression in follicular lymphoma. Blood. 82:1994–2004. 1993. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2020
Volume 56 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu Z, Li T, Zhang X, Zhang Z, Zhu D, Lin P, Tu S and Ren W: Molecular and clinical progress in follicular lymphoma lacking the t(14;18) translocation (Review). Int J Oncol 56: 7-17, 2020
APA
Zhu, Z., Li, T., Zhang, X., Zhang, Z., Zhu, D., Lin, P. ... Ren, W. (2020). Molecular and clinical progress in follicular lymphoma lacking the t(14;18) translocation (Review). International Journal of Oncology, 56, 7-17. https://doi.org/10.3892/ijo.2019.4917
MLA
Zhu, Z., Li, T., Zhang, X., Zhang, Z., Zhu, D., Lin, P., Tu, S., Ren, W."Molecular and clinical progress in follicular lymphoma lacking the t(14;18) translocation (Review)". International Journal of Oncology 56.1 (2020): 7-17.
Chicago
Zhu, Z., Li, T., Zhang, X., Zhang, Z., Zhu, D., Lin, P., Tu, S., Ren, W."Molecular and clinical progress in follicular lymphoma lacking the t(14;18) translocation (Review)". International Journal of Oncology 56, no. 1 (2020): 7-17. https://doi.org/10.3892/ijo.2019.4917