Review
Published: 2020-03-12
download
PDF

The broad landscape of follicular lymphoma: Part II

Department of Anatomic Pathology, St. Eugenio Hospital of Rome, Rome, Italy
Pathology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy
Surgical Oncology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy; Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
Pathology Unit, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Foggia, Foggia, Italy;
Pathology Unit, University of Cagliari, Cagliari, Italy;
Pathology Unit, University of Cagliari, Cagliari, Italy;
Pathology Unit, University of Siena, Siena, Italy
Pathology Unit, Ospedale di Ascoli Piceno, Ascoli, Italy
Hematology Unit, CREO, Azienda Ospedaliera di Perugia, University of Perugia, Perugia, Italy
Pathology Unit, Department of Medical, Surgical and Experimental Surgery, University of Sassari, Italy
General Surgery Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Guastalla, Reggio Emilia, Italy
Gastroenterology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy
Pathology Unit, Azienda Ospedaliera Santa Maria Terni, University of Perugia, Terni, Italy
follicular lymphoma B-cell centrocyte, centroblast

Abstract

Follicular lymphoma is a neoplasm derived from follicle center B cells, typically both centrocytes and centroblasts, in variable proportions according to the lymphoma grading. The pattern of growth may be entirely follicular, follicular and diffuse and rarely completely diffuse. It represents the second most common non-Hodgkin lymphoma, after diffuse large B-cell lymphoma and it is the most common low-grade mature B-cell lymphoma in Western countries. In the majority of cases, follicular lymphoma is a nodal tumor, occurring in adults and is frequently associated with the translocation t(14;18)(q32;q21)/IGH-BCL2. However, in recent years the spectrum of follicular lymphoma has expanded and small subsets of follicular lymphoma, which differ from common follicular lymphoma, have been identified and included in the current 2017 WHO classification. The aim of our review is to describe the broad spectrum of follicular lymphoma, pointing out that the identification of distinct clinicopathological.

Introduction

Follicular lymphoma (FL) is the most common low-grade mature germinal center B-cell lymphoma in Western countries, representing 20% to 30% of all non-Hodgkin lymphomas 1. The updated 2017 World Health Organization (WHO) Classification includes critical aspects about FL 1. In recent years, histological and clinical spectrum of germinal center derived B-cell neoplasms has expanded, leading to the conclusion that FL represents a far more heterogeneous entity than originally appreciated. In the previous review we illustrated FL variants encountered in diagnostic practice 2. Surgical pathologists and hematopathologists should be aware of the broad FL landscape, in order to avoid diagnostic pitfalls and get to more accurate diagnosis.

Although FL is mostly a nodal disease, it can involve primarily extranodal sites. In the current WHO classification, it is well recognized that FL arising at particular extranodal sites (i.e. duodenum, skin and testis) have clinicopathological features and outcomes different from conventional nodal FL 1. The skin and gastrointestinal (GI) tract are the most commonly involved extranodal sites. The site of involvement may affect disease prognosis 3. It is well recognized that stage I FL of the skin and duodenal-type FL have a significantly better outcome than nodal primary disease. Differently, stage I FLs of muscle, connective tissue and nervous system have significantly worse survival than nodal FLs. Other extranodal sites such as head and neck or respiratory system are not associated with worse survival.

Duodenal-type FL

Primary duodenal FL is a variant with distinctive biological and clinical features. It consists of polypoid lesions of the small bowel, more often in the second portion of duodenum 4. Patients are usually asymptomatic. The disease is localized and incidentally detected. Classically, small nodules involve mucosa and submucosa. The cellular composition recapitulates low-grade FL, with centrocytes and only rare centroblasts (Fig. 1). The cells are positive for CD10, CD20, BCL6 and BCL2 and carry t(14;18)/IGH/BCL2 (Fig. 2). Primary duodenal FL has an indolent clinical course, often without therapy. Radiotherapy can be used. Local recurrences can occur. Clinical evaluation and staging is essential to exclude systemic FL secondarily involving the bowel, which follows a more aggressive course 5.

In this setting, multiple lymphomatous polyposis (MLP) needs to be mentioned, which is a rare clinical presentation of primary gastrointestinal lymphomas. MLP patients typically present multiple polypoid lesions of different sizes throughout the gastrointestinal tract. Most cases are mantle cell lymphoma (MCL), others originate from mucosa-associated lymphoid tissue (MALT lymphoma) and a few cases are FLs 6-8.

Testicular FL

Primary testicular FL was initially described in children and subsequently in adults (Fig. 3) 9,10. It is usually a low stage disease (stage 1E). Histologically, it tends to show high-grade morphology (grade 3A) and the pattern of growth can be follicular or follicular and diffuse. Prominent fibrosis is often present. (Fig. 4). Testicular FL expresses germinal center (GC) markers (CD10, BCL6), but it usually lacks BCL2-protein expression and BCL2 gene rearrangements (Fig. 5). FL of the testis is typically a localized disease with an indolent clinical behavior and good prognosis. Optimal therapy for patients with low stage disease is not well defined. Most patients (children and adults) are treated with surgery plus anthracycline-containing chemotherapy, sometimes with central nervous system prophylaxis. In adults, primary FL of the testis needs to be distinguished from diffuse large B-cell lymphoma (DLBCL) (more commonly seen in the adult testis), which is a much more aggressive disease.

FL confined to the ovaries

FL arising primarily in the ovary is very rare and its clinicopathological features are not completely clear. Oznan et al. identified two main groups of FL arising in the ovary with divergent clinicopathological aspects 11. The first group included cases with high-grade histology (3A), negativity or weak positivity for BCL2 and absence of BCL2 translocation; this group frequently had low stage disease. The second group included cases with low histological grade, strong positivity for BCL2 protein and presence of IGH/BCL2 translocation; this group frequently had advanced stage disease. There is no clear evidence that the second group arises primarily in the ovary, as the disease is usually in an advanced stage. The first group (high-grade, low stage) includes cases with disease usually confined to the ovary. This latter group may represent true primary ovarian FL. The features of high-grade, low stage BCL2-negative FL resemble those reported in FL of the testis, which shows a favorable outcome. It is tempting to speculate that this group of ovarian FL might be related to either pediatric-type FL (PTFL) or testicular FL 11.

Primary cutaneous FL

Primary cutaneous FL (PCFL) is a low-grade lymphoma of follicle center B cells, without evidence of systemic/nodal involvement at time of diagnosis. It is the most common primary cutaneous B-cell lymphoma. It presents with localized plaques or nodules on the scalp, trunk and back and rarely on the legs; multifocal skin lesions can be present. PCFL involves the dermis, often extending into subcutis. The epidermis is spared with a grenz zone separating the epidermis from the underlying lymphoid proliferation (Fig. 6). Follicular and diffuse or totally diffuse pattern of growth may be present (Fig. 7) 12. Neoplastic follicles are usually closely packed, irregular and not polarized, tingible body macrophages are absent, and mantle zone is thin or absent. Neoplastic cells include centrocytes and centroblasts. Rarely, centrocytes may be spindle-shaped or show bizarre features with different sizes and shapes 12. Sclerosis and myxoid features may also be present in PCFL, particularly in the spindle cell variant 12. Reactive T cells can be prominent. PCFL with a diffuse pattern of growth is entirely composed of sheets of centrocytes and centroblasts. The neoplasm may show any grade, although high-grade cytology (grade 3) is seen quite frequently and differential diagnosis with DLBCL leg-type is mandatory. BCL6 is positive; CD10 is positive in cases with a follicular growth pattern, whereas it is often negative in cases with predominantly diffuse pattern (Fig. 8) 13. Although BCL2 is often reported as negative or weakly positive and t(14;18)/IGH-BCL2 is frequently absent, recent studies have identified BCL2 expression as well as the presence of BCL2 rearrangements in a proportion of PCFL 14,15. IRF4/MUM1 and FOXP1 are usually absent. Ki-67 shows a low proliferation index. Some PCFLs, mainly those composed of large cells and which need to be distinguished from DLBCL leg-type, can show high Ki-67. PCFL patients usually have an indolent disease, regardless of grade. Unlike nodal FL, PCFL should not be graded histologically, because grading does not seem to provide prognostic information. Surgical excision and local radiotherapy are treatments of choice for localized disease. Rare cases with multifocal skin lesions and extensive cutaneous disease require systemic therapy. Local recurrences are reported (20-30% of cases). When extracutaneous sites are involved, regional lymph nodes and bone marrow are usually affected 12. Transformation to DLBCL has been suggested by some studies 16.

Systemic FL secondarily involving the skin needs to be excluded. This scenario is challenging for pathologists, because careful clinical workup and staging are usually unavailable at time of skin biopsy. PCFL with diffuse pattern and numerous large centrocytes and centroblasts can be tricky to separate from DLBCL leg-type. The presence of centrocytes, follicular dendritic cells meshwork and numerous T cells argue in favor of PCFL. Sheets of large atypical centroblasts and/or immunoblasts support DLBCL leg-type. MUM1 and FOXP1 are usually absent in PCFL, differently from DLBCL leg-type 12.

Primary cutaneous marginal zone B-cell lymphoma (PCMZBCL) needs to be distinguished from PCFL (Fig. 9). It shows a nodular or diffuse infiltrate within the dermis and sometimes subcutis, with sparing of epidermis. At low power, PCMZBCL typically shows nodular infiltrates containing reactive GC surrounded by small- and medium-sized cells with pale cytoplasm (marginal-zone, centrocyte-like cells). Characteristically, PCMZBCL has a more heterogeneous cytological appearance, including small- to medium-sized mature centrocyte-like lymphocytes, monocytoid lymphocytes with abundant clear cytoplasm and plasma cells. Dutcher bodies may be identified. PCMZBCL lacks expression of BCL6, CD10, and CD5. Light chain restricted plasma cells are commonly found at the periphery of the lesion 17.

Primary cutaneous follicular helper T-cell lymphoma (PCFHTCL) is a relatively recently described lymphoma mimicking PCFL. Patients tend to be elderly, presenting with multiple nodules, papules and/or plaques, often involving the extremities 18. PCFHTCL displays a nodular architecture, syringotropism and includes numerous B cells. A careful examination reveals atypical T-cells with a follicular T-helper phenotype, including CD10 and BCL6 19. Systemic nodal follicular T-cell lymphoma may secondarily involve the skin; it can mimic follicular B-cell lymphoma.

Blastic plasmacytoid dendritic cell neoplasm is a rare myeloid neoplasm of immature plasmacytoid dendritic cells, commonly involving the skin. It can have a leukemic presentation or it can be limited to the skin, at least initially (Fig. 10). It follows an aggressive course, even in patients with skin-limited lesions. It may show a nodular growth pattern and include centrocyte-like cells. The expression of CD123, CD56, CD4, TCL1, CD2AP and CD303 helps with its correct classification 20.

Cutaneous reactive lymphoid hyperplasia may also mimic PCFL. In reactive hyperplasia, follicles show the typical outer mantle zone, encircling well polarized GC, as highlighted by Ki-67. Molecular studies (polymerase chain reaction - PCR) can help support PCFL diagnosis, examining immunoglobulin heavy-chain and light-chain genes and confirming clonality. The presence of clonality supports PCFL, if in the right clinicopathological context. However, pitfalls do exist. DNA may be of insufficient quality for molecular analysis, particularly in formalin-fixed, paraffin-embedded tissue. When DNA is of poor quality, false-negative results can occur 21. PCFL may not have a detectable monoclonal population and false-negative results are possible 22. In addition, when a B-cell population emerges as dominant clone in reactive lymphoid hyperplasia, false-positive results can occur 23.

Splenic FL

Primary splenic FL is very rare. To date, just a few studies on primary splenic FL have been performed 24. Its macroscopic appearance with multiple, small nodules can resemble splenic marginal zone lymphoma (Fig. 11). Histologically, the spleen shows a micronodular pattern, GC cytology and frequent marginal zone-like cells at nodules periphery (Fig. 12). Mollejo et al. reported the clinicopathological features of primary splenic FL subdividing it in 2 groups: the first resembling classical FL with the presence of t(14;18) and CD10 expression, which is usually diagnosed at advanced stage; the second, characterized by high grading, elevated proliferation index and BCL2 negativity, more often restricted to the spleen (Fig. 13) 24. Splenic FL shows some clinical features different from nodal FL. Hepatitis C virus (HCV)-positive status is significantly more common in patients with splenic FL. Ann Arbor stage III or IV and high-risk FLIPI (Follicular Lymphoma International Prognostic Index) are less common in splenic FL. The progression-free survival is worse in patients undergoing splenectomy without postoperative chemotherapy. These results suggest the spleen itself, as primary lesion, might affect the biological characteristics of FL. Splenic FL should probably be considered a distinct type of FL compared to nodal FL 25.

Rare extranodal sites involved by FL

Rarely, FL may involve uncommon extranodal sites such as peripheral nerves, muscle, peritoneum, dura, pancreas, conjunctiva and orbit 26-33.

Unusual phenotypes and molecular pitfalls in FL

BCL2-negative FL. BCL2 protein expression varies from 85-90% in grade 1-2 to less than 50% in grade 3 FL. BCL2 expression is related to the recurrent translocation t(14,18)(q32;q21) involving IGH and BCL2. BCL2-negative FL are explained by either true absence of t(14;18) or by mutation in BCL2 epitope usually recognized by clone 124 anti-BCL2 antibody. In these cases of BCL2 “pseudo-negative” FL, neoplastic follicles are immunoreactive using different anti-BCL2 antibodies such as clones E17 and/or SP66 34,35. Thus, the absence of BCL2 should not be interpreted as evidence against FL diagnosis, if other features are consistent with FL. Furthermore, the use of additional clones of anti-BCL2 antibody in the work-up of BCL2-negative FL is advisable.

CD10/BCL6 negative FL. A subset of FL, more frequently grade 3A, is CD10-negative and/or BCL6-negative. Recently, novel markers like Stathmin, GCET1, HGAL, and LMO2 have been introduced that can be useful in CD10 and/or BCL6-negative FL 36.

CD30 positive FL. A small percentage of FL, mostly grade 3, may contain sparse CD30-positive cells. This phenomenon is usually restricted to large centroblasts and/or to pleomorphic Hodgkin-Reed Sternberg (HRS)-like cells of grade 3 FL 37.

CD5 positive FL. CD5 is expressed by 5% of FL. CD5-positive FL can have the floral and/or diffuse patterns of growth. CD5 expression has been associated with higher International Prognostic Index (IPI), higher rate of transformation, and shorter progression-free survival 38.

IRF4/MUM1 positive FL. IRF4/MUM1 expression is detected in grade 3B FL as marker of late GC differentiation 39. Low to moderate IRF4/MUM1 expression may be observed even in low-grade FL. High IRF4/MUM1 expression has been recently reported to be predictive of poor outcome in low-grade FL 40.

The t(14;18)(q32;q21)/IGH-BCL2 translocation is present in common FL, although its frequency varies greatly depending on FL grading. The translocation t(14;18) is detected in up to 90% of low-grade FL, but in only 60-70% of grade 3A and 15-30% of grade 3B FL 41. Furthermore, BCL2 translocation variants such as t(2;18) and t(18;22) have been described. FL with BCL6 translocation represents 10-15% of cases, more frequently grade 3A and 3B. These FL strongly express BCL6, but are quite often BCL2 and/or CD10 negative 42.

The updated WHO classification recognizes the category of high-grade B-cell lymphoma (HGBCL) with MYC and BCL2 and/or BCL6 rearrangements, so called double-hit (DH) or triple-hit (TH) lymphomas 1. Occasionally, “de novo” low-grade or grade 3 FL may carry MYC and BCL2 and/or BCL6 gene rearrangements, but should not be classified as HGBCL, unless undergoing a clear-cut transformation. The prognostic significance of concurrent MYC and BCL2 or BCL6 rearrangements in otherwise typical FL is an open question. Some studies report an aggressive course, but better response to more intensive regimens, while others show a behavior similar to FL lacking MYC rearrangement 43-45.

Elucidation of the clinicopathological features of NOTCH-mutant FL. NOTCH1 and NOTCH2 have been recently reported in several B cell lymphoma. The role of these mutations in FL is not known. A recent study identified NOTCH1 and NOTCH2 mutations in 6.3% of FL. NOTCH-mutated FL showed lower frequency of t(14;18), higher incidence of splenic involvement and female predominance. Furthermore, transformation was more frequently identified in NOTCH-mutated FL than in wild-type cases. These results indicate NOTCH mutations are uncommon in FL, but may occur in a subset of cases with distinctive features 46.

Grade 3B FL

The number of centroblasts is the key feature for FL grading. Grade 3B is a FL with a purely follicular growth pattern, composed only by centroblasts (Fig. 14). Pure 3B FL is rare and clinical data on this enigmatic entity remain scarce 39. Most 3B FLs focally contain diffuse areas, therefore, deserving the diagnosis of DLBCL 47. Furthermore, grade 3B rarely coexists with grade 1-2 or 3A, suggesting a divergent pathogenesis 48. Grade 3B FL is generally CD10-negative and IRF4/MUM1-positive (Fig. 15). The translocation t(14;18)(q32;q21) juxtaposing the IGH and BCL2 genes is rare in pure 3B FL (13%), despite expressing BCL2 protein in 69% of cases. BCL6 rearrangement occurs rarely in pure 3B FL, whereas increased TP53 expression is rather common (31%). Grade 3B FL is still an evolving subclass. Physicians should understand its aggressive nature, requiring timely attention, compared with grade 3A. In many aspects 3B FL resembles de novo DLBCL. Some studies suggest it may represent a morphological variant of DLBCL with a follicular pattern of growth 39. Histology, immunophenotypic profile and chromosomal aberrations of pure 3B FL resemble DLBCL, particularly non-GCB type. It is widely accepted 3B FL is distinct from other types of FL and it is intriguing to speculate it may represent a follicular growing variant of DLBCL.

Transformation of nodal FL

It is well recognized that clinical aggressiveness and risk of transformation to DLBCL increase proportionally to the number of centroblasts and proliferative fraction. Transformation or progression occurs in 30% of FL. The current WHO criteria for transformed FL include a diffuse pattern of growth with centroblasts > 15/HPF (grade 3) 1. In other words, the presence of grade 3 cytology in a diffuse pattern constitutes a DLBCL.

Currently, transformed FL is classified as DLBCL, or high-grade B-cell lymphoma (HGBCL) with MYC, BCL2 and/or BCL6 rearrangements or HGBCL not otherwise specified (in absence of MYC, BCL2 and/or BCL6 rearrangements). The HGBCL category has variable morphology, including DLBCL, Burkitt lymphoma, and/or “blastoid” morphology. “Burkitt-like” cases are reminiscent of both DLBCL and Burkitt lymphoma, not fulfilling diagnostic criteria for either entity. Cases with so-called “blastoid morphology” show diffuse cohesive sheets of monotonous, small to medium-sized cells, high proliferation index, and starry-sky pattern. The cells have round nuclei with finely dispersed chromatin, inconspicuous nucleoli and a small rim of cytoplasm. These cells resemble lymphoblasts or the blastoid variant of MCL. Staining for TdT, Cyclin D1 and SOX11 should be performed.

More rarely, FL may transform into Hodgkin lymphoma, plasmablastic lymphoma, histiocytic sarcoma (HS) or precursor B cell lymphoblastic leukemia/lymphoma 49-52.

Lymphoblastic-type transformation of FL is a rare event with a poor outcome. It has to be differentiated from “de novo” FL with blastoid features, which has the typical FL phenotype and genetic abnormalities, without TdT expression. To avoid any confusion, the current WHO classification recommends the term “transformed FL of lymphoblastic type, TdT positive”.

Recent studies suggested that transformation might occur in early neoplastic progenitors rather than in later subclones 53. Thus, the phenomenon of transformation could be explained by a divergent evolution from a common precursor which was the founder cell of initial FL, and then evolved into DLBCL, histiocyte sarcoma, or lymphoblastic B-cell neoplasia 54.

Clonality studies are useful; matching rearrangements between initial FL and subsequent high grade lymphoma confirm a clonal relationship.

Composite FL

Composite lymphoma (CL) represents a fascinating process. It consists of two or more morphologically and immunophenotypically distinct lymphomas within the same anatomic site 55,56. Its incidence ranges from 1 to 4.7% of total lymphomas, although CL may be more common than previously thought 57,58. CL can arise synchronously or metachronously and can be clonally related or not. Regardless of the distinctive histology of the different components, in some cases the components are clonally related, whereas in others they are clonally unrelated, representing the “collision” of clonally unrelated tumors. With the advent of molecular analysis, it became clear that, in a subset of cases, CL components share a common clonal origin, suggesting derivation from a common precursor cell 59,60.

An adequate sampling is required to establish CL diagnosis. CL can be composed of FL and MCL, FL and chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), FL and nodal marginal zone lymphoma (NMZL), FL and nodular lymphocyte predominance Hodgkin lymphoma (NLPHL), FL and classic Hodgkin lymphoma (CHL). Cases of FL associated with DLBCL, HGBCL, Burkitt lymphoma and B-lymphoblastic lymphoma/leukemia have to be excluded, because they represent high-grade transformation.

CL with FL and MCL have been rarely reported. FL component is typically low-grade, BCL2 positive and harbors the t(14;18) translocation. The MCL component shows a diffuse or in situ mantle-zone growth pattern, it is CCND1 positive and harbors the t(11;14) translocation 61. Morphologically, the nodal architecture is intact and reactive follicles are mainly distributed in the cortex. The mantle zone is preserved and CCND1 positive cells are often restricted to the mantle zone 62. Some studies suggest FL and MCL are clonally related, originating from the same preneoplastic clone 59,63,64.

CL with FL and CLL/SLL is extremely rare (Fig. 16) 65. An interesting study of Boiocchi et al. supported the notion that composite low-grade B-cell lymphomas are usually biclonal 66. Another recent study, reporting the largest case series of composite CLL/SLL and FL did not perform microdissection, so that the relationship between the two components cannot be definitively determined 65.

Recent examples of composite FL and NMZL, studied by PCR sequencing of IGH from microdissected NMZL and FL components, showed different sequences in the CDR3 region, suggesting the presence of two different clones 67.

Rarely, composite FL and NLPHL have been described, but the clonal relationship was not established, due to insufficient tissue for laser capture microdissection 68,69. Composite CHL and FL have been reported, in some of which the CHL and FL components were clonally related. CHL may display the translocation t(14;18), suggesting a common origin (common B-cell precursor) of CHL and FL components. In a series of 19 composite cases involving CHL and other non-Hodgkin lymphomas, a shared clonality was demonstrated in 12/19 (63%) cases 58,70,71.

T-cell lymphoma associated with low-grade B-cell lymphoma is very rare. A composite FL and T-cell lymphoma is rarely reported 72,73. The genomic aberration may have occurred in an early lymphoid progenitor which underwent divergent evolution via additional genomic alterations, resulting in heterogeneous subclones and eventually T-cell and B-cell neoplasms 72.

Occasionally, small innocuous aggregates of Langherans cells are identified within FL. Histiocytic and Langherans cell neoplasms occurring synchronously or sequentially in FL patients have been reported. FL and Langherans cell neoplasms or histiocytic sarcoma often share a common cell precursor (clonally related) 51,74.

CL diagnosis is challenging, requiring careful interpretation of morphology, immunohistochemistry and fluorescence in situ hybridization (FISH) analysis as well as flow cytometry, particularly when both components show identical immunoglobulin light chain restrictions and/or overlapping immunophenotypic features. A handful of CL have been reported, few of which have been characterized in terms of clonal relationships. IGH gene rearrangement analysis is critical to demonstrate the clonal relationship. It is recommended to use not only morphology, immunohistochemistry and FISH, but also PCR or next-generation sequencing (NGS) of the IGH and T-cell receptor gene rearrangements. Molecular studies are proving to be invaluable in CL workup. FISH, immunoglobulin rearrangement and sequencing as well as NGS technology can be improved by tissue microdissection. IGH analysis on whole tissue sections may not be helpful and laser capture microdissection is necessary to purify, or enrich individual components, in order to allow an interpretable gene rearrangement analysis. Thus, the power of microdissection coupled with molecular analysis needs to be considered. Recently, the concept is emerging that CL may represent different phenotypes of an identical shared common progenitor 71.

The analysis of additional CL cases is necessary to further investigate the clonal relationship between the individual components and to get better insights into CL pathogenesis.

Bone marrow involvement by FL

Usually, bone marrow trephine biopsy is performed for FL staging. Bone marrow involvement is quite common, occurring in 80% of FL patients. Typically, lymphoma is aligned along the trabecular bone (paratrabecular pattern), although interstitial and/or nodular patterns may be seen (Fig. 17). Rarely, bone marrow is extensively involved by FL (Fig. 18) 75. Grading is not recommended on bone marrow biopsy. In absence of previous rituximab therapy, CD20 is sufficient to reveal even subtle bone marrow infiltration, whereas CD10 and BCL6 are typically downregulated or may be totally negative. BCL2 staining is not useful, and does not add any further information. Furthermore, BCL2 is expressed by many other indolent low-grade B-cell lymphomas. Sometimes, a nodal transformed DLBCL coexists with low-grade FL in bone marrow, representing the so-called “discordant” lymphoma 76.

Approach to histopathological diagnosis of FL by core needle biopsy

Correct lymphoma classification is the best way to obtain relevant information for treatment and outcomes. The criteria for FL diagnosis and, by extension, the most appropriate therapeutic strategies are based largely on histologic evaluation of surgically excised specimens. Nonetheless, recently, an increasing reliance on core needle biopsy (CNB) of lymph nodes is evident. In many institutions, CNB is the primary diagnostic procedure in the suspect of lymphoma. Several studies on the effectiveness of CNB suggested that CNB yields an adequate diagnosis for treatment decision in about 65% to 75% of cases 77-80.

The reasons of the increasing popularity of this procedure are briefly summarized below. One of the most important considerations leading to CNB over excisional biopsy is urgency. However, in 25% of cases, CNB fails to yield an actionable diagnosis, further delaying therapy (Tab. I).

Since CNB can give only partial information, excisional biopsy of the lymph node should be performed, whenever possible. CNB has limitations, which is not particularly surprising, given how critical the architectural pattern is in FL diagnosis (Tab. II). Histological pattern, grading, immunohistochemical interpretation, including proliferative index, as well as detection of areas of transformation are common dilemmas, as many samples do not contain the recommended 10 follicles. Accurate grading may be very difficult on CNB. The National Comprehensive Cancer Network (NCCN) clinical practice guidelines in oncology are quite explicit, regarding the preference of excisional biopsies at the time of initial diagnosis, whenever feasible 77,80,81. Despite the evolution of diagnostic methodologies, the use of ancillary techniques only occasionally compensates the loss of diagnostic specificity due to limited sampling.

The current WHO classification states “accurate grading cannot be performed on fine-needle aspiration and may be difficult on core needle biopsy. Therefore, an excisional biopsy is recommended for primary diagnosis”.

Conclusion

Under the broad heading of FL, diseases with different clinicopathological features are included. Diverse molecular pathways are probably associated with different clinical features and outcomes.

Figures and tables

Figure 1.A Ileum. Follicular lymphoma, duodenal-type. Giemsa, 100x; B Ileum. Follicular lymphoma, duodenal-type. Giemsa, 200x; C Ileum. Follicular lymphoma, duodenal-type. Giemsa, 400x.

Figure 2.A Ileum. Follicular lymphoma, duodenal-type. CD20 immunostaining, 100x; B Ileum. Follicular lymphoma, duodenal-type. BCL6 immunostaining, 100x; C Ileum. Follicular lymphoma, duodenal-type. BCL2 immunostaining, 100x; D Ileum. Follicular lymphoma, duodenal-type. KI67/MIB1 immunostaining, 100x.

Figure 3.Testis. Primary follicular lymphoma of the testis. Gross appearance. Courtesy of Dr. A. Pireddu, Anatomia Patologica, Ospedale di Città di Castello, Perugia, Italy.

Figure 4.A Follicular lymphoma of the testis. HE, 100x; B Follicular lymphoma of the testis. HE, 200x; C Follicular lymphoma of the testis. HE, 400x.

Figure 5.A Follicular lymphoma of the testis. CD10 immunostaining, 400x; B Follicular lymphoma of the testis. BCL6 immunostaining, 400x; C Follicular lymphoma of the testis. BCL2 immunostaining, 400x; D Follicular lymphoma of the testis. Ki-67/MIB1 immunostaining, 400x.

Figure 6.A Primary cutaneous follicular lymphoma. HE, 200x; B Primary cutaneous follicular lymphoma. HE, 400x; C Primary cutaneous follicular lymphoma. CD20 immunostaining, 100x; D Primary cutaneous follicular lymphoma. BCL6 immunostaining, 100x; E Primary cutaneous follicular lymphoma. BCL2 immunostaining, 100x; F Primary cutaneous follicular lymphoma. Ki-67/MIB1 immunostaining, 400x.

Figure 7.A Primary cutaneous follicular lymphoma, follicular pattern of growth. HE, 100x; B Primary cutaneous follicular lymphoma, follicular pattern of growth. HE, 200x; C Primary cutaneous follicular lymphoma, follicular pattern of growth. HE, 400x; D Primary cutaneous follicular lymphoma, diffuse pattern of growth. HE, 40x; E Primary cutaneous follicular lymphoma, diffuse pattern of growth. Large-sized neoplastic cells. HE, 400x.

Figure 8.A Primary cutaneous follicular lymphoma, diffuse pattern of growth. CD10 immunostaining, 100x; B Primary cutaneous follicular lymphoma, diffuse pattern of growth. Large-sized neoplastic cells. BCL6 immunostaining, 200x; C Primary cutaneous follicular lymphoma, diffuse pattern of growth. Large-sized neoplastic cells. LMO2 immunostaining, 200x; D Primary cutaneous follicular lymphoma, diffuse pattern of growth. Large-sized neoplastic cells. BCL2 immunostaining, 400x; E Primary cutaneous follicular lymphoma, diffuse pattern of growth. Large-sized neoplastic cells. FOXP1 immunostaining, 400x; F Primary cutaneous follicular lymphoma, diffuse pattern of growth. Large-sized neoplastic cells. KI-67/MIB1 immunostaining, 200x.

Figure 9.A Skin. Marginal lymphoma. Small, medium-sized lymphoid cells and few mature plasma cells. HE, 200x; B Skin. Marginal lymphoma. Monotypic mature plasma cell component. Kappa immunostaining, 200x; C Skin. Marginal lymphoma. Monotypic mature plasma cell component. Lambda immunostaining, 200x.

Figure 10.Skin. Blastic plasmacytoid dendriitic cell neoplasm. “Centrocyte-like” features of the neoplastic elements. HE, 400x.

Figure 11.A Follicular lymphoma of the spleen, gross appearance. Courtesy of Dr. Marco Toraldo, Anatomia Patologica, Ospedale di Foligno; B Follicular lymphoma of the spleen, gross appearance. Courtesy of Dr. Marco Toraldo, Anatomia Patologica, Ospedale di Foligno.

Figure 12.A Follicular lymphoma of the spleen, follicular pattern of growth. HE, 100x; B Follicular lymphoma of the spleen. Numerous centrocytes and few centroblasts. HE, 400x.

Figure 13.A Follicular lymphoma of the spleen. CD3-negative neoplastic elements, 100x; B Follicular lymphoma of the spleen. CD20-positive neoplastic elements, 100x; C Follicular lymphoma of the spleen. CD10-positive neoplastic elements, 100x; D Follicular lymphoma of the spleen. BCL6-positive neoplastic elements, 100x; E Follicular lymphoma of the spleen. BCL2-positive neoplastic elements, 100x; F Follicular lymphoma of the spleen. Proliferative index of neoplastic elements (KI-67/MIB1 15-20%), 100x.

Figure 14.A Lymph node. Follicular lymphoma 3B. Giemsa, 40x; B Lymph node. Follicular lymphoma 3B. Giemsa, 100x; C Lymph node. Follicular lymphoma 3B. Giemsa, 400x.

Figure 15.A Lymph node. Follicular lymphoma 3B. CD20 immunostaining, 40x; B Lymph node. Follicular lymphoma 3B. CD10 immunostaining, 200x; C Lymph node. Follicular lymphoma 3B. BCL6 immunostaining, 200x; D Lymph node. Follicular lymphoma 3B. BCL2 immunostaining, 400x; E Lymph node. Follicular lymphoma 3B. LMO2 immunostaining, 100x; F Lymph node. Follicular lymphoma 3B. KI-67/MIB1 immunostaining, 100x.

Figure 16.A Lymph node. Composite lymphoma: Follicular lymphoma and small lymphocytic lymphoma/chronic lymphocytic leukaemia. Giemsa, 100x; B Lymph node. Composite lymphoma: Follicular lymphoma and small lymphocytic lymphoma/chronic lymphocytic leukaemia. Giemsa, 200x; C Lymph node. Composite lymphoma: Follicular lymphoma and small lymphocytic lymphoma/chronic lymphocytic leukaemia. Giemsa, 400x; D Lymph node. Composite lymphoma: Follicular lymphoma and small lymphocytic lymphoma/chronic lymphocytic leukaemia. CD5 immunostaining. 100x; E Lymph node. Composite lymphoma: Follicular lymphoma and small lymphocytic lymphoma/chronic lymphocytic leukaemia. CD23 immunostaining. 100x; F Lymph node. Composite lymphoma: Follicular lymphoma and small lymphocytic lymphoma/chronic lymphocytic leukaemia. BCL2 immunostaining. 100x.

Figure 17.A Bone marrow. Follicular lymphoma, paratrabecular pattern of growth. HE, 100x; B Bone marrow. Follicular lymphoma, paratrabecular pattern of growth. HE, 400x; C Bone marrow. Follicular lymphoma, paratrabecular pattern of growth. CD20 immunostaining, 100x.

Figure 18.A Bone marrow. Follicular lymphoma, diffuse pattern of growth. HE, 100x; B Bone marrow. Follicular lymphoma, diffuse pattern of growth. HE, 200x.

1 Diagnostic urgency
2 Sparing surgical procedure to the patient [patient compliance]
3 Risk of complications minimized [little morbidity]
4 Lymph nodes difficult to be removed can be sampled [deep-seated lymph nodes]
5 75% less expensive than excisional lymph node biopsy [cost-effectiveness]
6 Excluding non-hematopoietic tumors
7 Staging patients with known lymphoma
8 Assessment for relapse of lymphoma [change in the histologic findings; transformation]
Table I.Advantages of core needle biopsy.
1 Small specimen size
2 Sampling error
3 Artifactual distortion of morphologic features
4 Adequacy of tissue for ancillary studies [exhaustion of the paraffin block]
Table II.Disadvantages of core needle biopsy.

References

  1. Swerdlow SH, Campo E, Harris NL. Revised 4th Edition WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. IARC Press: Lyon, France; 2017.
  2. Fratoni S, Zanelli M, Zizzo M. The broad landscape of follicular lymphoma: Part I. Pathologica. 2020; 112:1-16. DOI
  3. Shastri A, Janakiram M, Mantzaris I. Sites of extranodal involvement are prognostic in patients with stage 1 follicular lymphoma. Oncotarget. 2017; 8:78410-8. DOI
  4. Misdraji J, Harris NL, Hasserjian RP. Primary follicular lymphoma of the gastrointestinal tract. Am J Surg Pathol. 2011; 35:1255-63. DOI
  5. Damaj G, Verkarre V, Delmer A. Primary follicular lymphoma of the gastrointestinal tract: a study of 25 cases and a literature review. Ann Oncol. 2003; 14(4):623-9. DOI
  6. Sakata Y, Iwakiri R, Sakata H. Primary gastrointestinal follicular center lymphoma resembling multiple lymphomatous polyposis. Dig Dis Sci. 2001; 46:567-70. DOI
  7. Huang WT, Hsu YH, Yang SF. Primary gastrointestinal follicular lymphoma: a clinicopathologic study of 13 cases from Taiwan. J Clin Gastroenterol. 2008; 42:997-1002. DOI
  8. Zanelli M, Tioli C, Mengoli MC. Marginal-zone lymphoma: a rare presentation with multiple intestinal polyps. Clin Res Hepatol. 2019; 43(2):117-9. DOI
  9. Pileri SA, Sabattini E, Rosito P. Primary follicular lymphoma of the testis in childhood: an entity with peculiar clinical and molecular characteristics. J Clin Pathol. 2002; 55:684-8. DOI
  10. Bacon CM, Ye H, Diss TC. Primary follicular lymphoma of the testis and epididymis in adults. Am J Surg Pathol. 2007; 31:1050-8. DOI
  11. Ozsan N, Bedke BJ, Law ME. Clinicopathologic and genetic characterization of follicular lymphomas presenting in the ovary reveals 2 distinct subgroups. Am J Surg Pathol. 2011; 35:1691-9. DOI
  12. Cerroni L.. Skin Lymphoma: The Illustrated Guide. Wiley Blackwell: Hoboken, NJ; 2014.
  13. Servitje O, Climent F, Colomo L. Primary cutaneous vs secondary cutaneous follicular lymphomas: A comparative study focused on BCL2, CD10, and t(14;18) expression. J Cutan Pathol. 2019; 46:182-9. DOI
  14. Vergier B, Belaud-Rotureau MA, Benassy MN. Neoplastic cells do not carry bcl2-JH rearrangements detected in a subset of primary cutaneous follicle center B-cell lymphomas. Am J Surg Pathol. 2004; 28:748-55. DOI
  15. Szablewski V, Ingen-Housz-Oro S, Baia M. Primary Cutaneous Follicle Center Lymphomas Expressing BCL2 Protein Frequently Harbor BCL2 Gene Break and May Present 1p36 Deletion: A Study of 20 Cases. Am J Surg Pathol. 2016; 40:127-36. DOI
  16. Tsang HC, Mathew S, Magro CM. An aggressive primary cutaneous follicle center lymphoma with c-MYC translocation and CDKN2A (9p21) deletion: a case report and review of the literature. Am J Dermatopathol. 2017; 39:e44-9. DOI
  17. Swerdlow SH.. Cutaneous marginal zone lymphomas. Semin Diagn Pathol. 2017; 34:76-84. DOI
  18. Battistella M, Beylot-Barry M, Bachelez H. Primary cutaneous follicular helper T-cell lymphoma: a new subtype of cutaneous T-cell lymphoma reported in a series of 5 cases. Arch Dermatol. 2012; 148:832-9. DOI
  19. Wang JY, Nguyen GH, Ruan J. Primary cutaneous follicular helper t-cell lymphoma: a case series and review of the literature. Am J Dermatopathol. 2017; 39:374-83. DOI
  20. Cota C, Vale E, Viana I. Cutaneous manifestations of blasticplasmacytoid dendritic cell neoplasm-morphologic and phenotypic variability in a series of 33 patients. Am J Surg Pathol. 2010; 34:75-87. DOI
  21. Evans PA, Pott Ch, Groenen PJ. Significantly improved PCR-based clonality testing in B-cell malignancies by use of multiple immunoglobulin gene targets. Report of the BIOMED-2 concerted action BHM4-CT98-3936. Leukemia. 2007; 21:207-14. DOI
  22. Schafernak KT1, Variakojis D, Goolsby CL. Clonality assessment of cutaneous B-cell lymphoid proliferations: a comparison of flow cytometry immunophenotyping, molecular studies, and immunohistochemistry/in situ hybridization and review of the literature. Am J Dermatopathol. 2014; 36:781-95. DOI
  23. Gulia A, Saggini A, Wiesner T. Clinicopathologic features of early lesions of primary cutaneous follicle center lymphoma, diffuse type: implications for early diagnosis and treatment. J Am Acad Dermatol. 2011; 65:991-1000. DOI
  24. Mollejo M, Rodríguez-Pinilla MS, Montes-Moreno S. Splenic follicular lymphoma: clinicopathologic characteristics of a series of 32 cases. Am J Surg Pathol. 2009; 33:730-8. DOI
  25. Shimono J, Miyoshi H, Kamimura T. Clinicopathological features of primary splenic follicular lymphoma. Ann Hematol. 2017; 96:2063-70. DOI
  26. Umeda M, Kondo T, Nishikori M. A case of neurolymphomatosis caused by follicular lymphoma successfully treated with bendamustine. Clin Case Rep. 2015; 4:23-5. DOI
  27. Martins F, Stalder G, Van Der Gucht A. Intramuscular Follicular Lymphoma. Clin Nucl Med. 2018; 43:682-4. DOI
  28. Cunningham N, Ffrench-Constant S, Planche K. Peritoneal lymphomatosis: a rare presentation of follicular lymphoma mimicking peritoneal carcinomatosis. BMJ Case Rep. 2015; 2015:bcr2014207136. DOI
  29. Tandon R, Mukherjee U, Abrari A. Primary dural follicular lymphoma masquerading as meningioma: a case report. Br J Neurosurg. 2012; 26:905-6. DOI
  30. Sallapan S, Abu Bakar NZ, Jarmin R. Primary follicular lymphoma of the pancreas: A rare tumour mimicking pancreatic carcinoma. Malays J Pathol. 2018; 40:359-71.
  31. Gaffar M, Thebpatiphat N, Przygodzki R. Primary follicular lymphoma of the conjunctiva in a 6-year-old child. J AAPOS. 2010; 14:538-40. DOI
  32. Prasad GL, Hegde A, Menon G. Primary orbital follicular lymphoma: a case report and review. J Clin Diagn Res. 2017; 11:XD01-XD03. DOI
  33. Rasmussen PK, Ralfkiaer E, Prause JU. Follicular lymphoma of the ocular adnexal region: a nation-based study. Acta Ophthalmol. 2015; 93:184-9. DOI
  34. Schraders M, de Jong D, Kluin P. 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. 2005; 205:329-35. DOI
  35. Adam P, Baumann R, Schmidt J. The BCL2 E17 and SP66 antibodies discriminate 2 immunophenotypically and genetically distinct subgroups of conventionally BCL2-”negative” grade 1/2 follicular lymphomas. Hum Pathol. 2013; 44:1817-26. DOI
  36. Younes SF, Beck AH, Ohgami RS. The efficacy of HGAL and LMO2 in the separation of lymphomas derived from small B cells in nodal and extranodal sites, including the bone marrow. Am J Clin Pathol. 2011; 135:697-708. DOI
  37. Xerri L, Dirnhofer S, Quintanilla-Martinez L. The heterogeneity of follicular lymphomas: from early development to transformation. Virchows Arch. 2016; 468:127-39. DOI
  38. Li Y, Hu S, Zuo Z. CD5-positive follicular lymphoma: clinicopathologic correlations and outcome in 88 cases. Mod Pathol. 2015; 28:787-98. DOI
  39. Horn H, Schmelter C, Leich E. Follicular lymphoma grade 3B is a distinct neoplasm according to cytogenetic and immunohistochemical profiles. Haematologica. 2011; 96:1327-34. DOI
  40. Xerri L, Bachy E, Fabiani B. Identification of MUM1 as a prognostic immunohistochemical marker in follicular lymphoma using computerized image analysis. Hum Pathol. 2014; 45:2085-93. DOI
  41. Bosga-Bouwer AG, van Imhoff GW, Boonstra R. 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. 2003; 101:1149-54. DOI
  42. Díaz-Alderete A, Doval A, Camacho F. Frequency of BCL2 and BCL6 translocations in follicular lymphoma: relation with histological and clinical features. Leuk Lymphoma. 2008; 49:95-101. DOI
  43. Miao Y, Hu S, Lu X. Double-hit follicular lymphoma with MYC and BCL2 translocations: a study of 7 cases with a review of literature. Hum Pathol. 2016; 58:72-7. DOI
  44. Yoshida M, Ichikawa A, Miyoshi H. Clinicopathological features of double-hit B-cell lymphomas with MYC and BCL2, BCL6 or CCND1 rearrangements. Pathol Int. 2015; 65:519-27. DOI
  45. Miyaoka M, Kikuti YY, Carrreras J. Clinicopathological and genomic analysis of double-hit follicular lymphoma: comparison with high-grad B-cell lymphoma with MYC and CL2 and/or BCL6 rearrangements. Mod Pathol. 2018; 31:313-26. DOI
  46. Karube K, Martínez D, Royo C. Recurrent mutations of NOTCH genes in follicular lymphoma identify a distinctive subset of tumours. J Pathol. 2014; 234:423-30. DOI
  47. Salaverria I, Siebert R. Follicular lymphoma grade 3B. Best Pract Res Clin Haematol. 2011; 24:111-9. DOI
  48. Koch K, Hoster E, Ziepert M. Clinical, pathological and genetic features of follicular lymphoma grade 3A: a joint analysis of the German low-grade and high-grade lymphoma study groups GLSG and DSHNHL. Ann Oncol. 2016; 27:1323-9. DOI
  49. Scaramucci L, Perrotti A, Niscola P. Hodgkin disease subsequent to follicular lymphoma on maintenance rituximab. Leuk Lymphoma. 2007; 48:1878-80. DOI
  50. Martinez D, Valera A, Perez NS. Plasmablastic transformation of low-grade B-cell lymphomas: report on 6 cases. Am J Surg Pathol. 2013; 37:272-81. DOI
  51. Feldman AL, Arber DA, Pittaluga S. Clonally related follicular lymphomas and histiocytic/dendritic cell sarcomas: evidence for transdifferentiation of the follicular lymphoma clone. Blood. 2008; 111:5433-9. DOI
  52. Geyer JT, Subramaniyam S, Jiang Y. Lymphoblastic transformation of follicular lymphoma: a clinicopathologic and molecular analysis of 7 patients. Hum Pathol. 2015; 46:260-71. DOI
  53. Green MR, Alizadeh AA. Common progenitor cells in mature B-cell malignancies: implications for therapy. Curr Opin Hematol. 2014; 21:333-40. DOI
  54. Okosun J, Bödör C, Wang J. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat Genet. 2014; 46:176-81. DOI
  55. Gonzalez CL, Medeiros LJ, Jaffe ES. Composite lymphoma. A clinicopathologic analysis of nine patients with Hodgkin’s disease and B-cell non-Hodgkin’s lymphoma. Am J Clin Pathol. 1991; 96:81-9. DOI
  56. Kim H.. Composite lymphoma and related disorders. Am J Clin Pathol. 1993; 99:445-51. DOI
  57. Thirumala S, Esposito M, Fuchs A. An unusual variant of composite lymphoma: a short case report and review of the literature. Arch Pathol Lab Med. 2000; 124:1376-8.
  58. Maeshima AM, Taniguchi H, Nomoto J. Clinicopathological features of classical Hodgkin lymphoma in patients ≥ 40 years old, with special reference to composite cases. Jpn J Clin Oncol. 2015; 45:921-8. DOI
  59. Wang S, Tzankov A, Xu-Monette ZY. Clonally related composite follicular lymphoma and mantle cell lymphoma with clinicopathologic features and biological implications. Hum Pathol. 2013; 44:2658-67. DOI
  60. Schneider S, Crescenzi B, Schneider M. Subclonal evolution of a classical Hodgkin lymphoma from a germinal center B-cell-derived mantle cell lymphoma. Int J Cancer. 2014; 134:832-43. DOI
  61. Subtil A, Xu Z. Follicular lymphoma with composite in situ mantle cell neoplasia. Blood. 2019; 133:2460. DOI
  62. Carvajal-Cuenca A, Sua LF, Silva NM. In situ mantle cell lymphoma: clinical implications of an incidental finding with indolent clinical behavior. Haematologica. 2012; 97:270-8. DOI
  63. Tsang P, Pan L, Cesarman E. A distinctive composite lymphoma consisting of clonally related mantle cell lymphoma and follicle center cell lymphoma. Hum Pathol. 1999; 30:988-92. DOI
  64. Taverna J, Nair A, Yun S. A rare presentation of in situ mantle cell lymphoma and follicular lymphoma: a case report and review of the literature. Case Rep Hematol. 2014; 2014:145129. DOI
  65. Jelloul FZ, Chen QH, Yang T. Composite Small Lymphocytic Lymphoma/Chronic Lymphocytic Leukemia and Follicular Lymphoma: A Clinicopathological Study of Six Cases. Int J Surg Pathol. 2018; 26:135-44. DOI
  66. Boiocchi L, Witter RE, He B. Composite chronic lymphocytic leukemia/small lymphocytic lymphoma and follicular lymphoma are biclonal lymphomas: a report of two cases. Am J Clin Pathol. 2012; 137:647-59. DOI
  67. Miyaoka M, Kikuchi T, Carreras J. Composite follicular lymphoma and CD5-positive nodal marginal zone lymphoma. J Clin Exp Hematop. 2016; 56:55-8. DOI
  68. Dargent JL, Lespagnard L, Meiers I. Composite follicular lymphoma and nodular lymphocyte predominance Hodgkin’s disease. Virchows Arch. 2005; 447:778-80. DOI
  69. O’Neill JP, Quinn F, Dowling A. Composite t(14;18)-Negative follicular lymphoma and nodular lymphocyte-predominant hodgkin lymphoma. Case Rep Hematol. 2018; 2018:4312594. DOI
  70. Küppers R, Sousa AB, Baur AS. Common germinal-center B-cell origin of the malignant cells in two composite lymphomas, involving classical Hodgkin’s disease and either follicular lymphoma or B-CLL. Mol Med. 2001; 7:285-92.
  71. Küppers R, Dührsen U, Hansmann ML. Pathogenesis, diagnosis, and treatment of composite lymphomas. Lancet Oncol. 2014; 15:435-46. DOI
  72. Wang E, Papavassiliou P, Wang AR. Composite lymphoid neoplasm of B-cell and T-cell origins: a pathologic study of 14 cases. Hum Pathol. 2014; 45:768-84. DOI
  73. Tanaka J, Su P, Luedke C. Composite lymphoma of follicular B-cell and peripheral T-cell types with distinct zone distribution in a 75-year-old male patient: a case study. Hum Pathol. 2018; 76:110-6. DOI
  74. West DS, Dogan A, Quint PS. Clonally related follicular lymphomas and Langerhans cell neoplasms: expanding the spectrum of transdifferentiation. Am J Surg Pathol. 2013; 37:978-86. DOI
  75. Torlakovic E, Torlakovic G, Brunning RD. Follicular pattern of bone marrow involvement by follicular lymphoma. Am J Clin Pathol. 2002; 118:780-6. DOI
  76. Sovani V, Harvey C, Haynes AP. Bone marrow trephine biopsy involvement by lymphoma: review of histopathological features in 511 specimens and correlation with diagnostic biopsy, aspirate and peripheral blood findings. J Clin Pathol. 2014; 67:389-95. DOI
  77. Frederiksen JK, Sharma M, Casulo C. Systematic review of the effectiveness of fine-needle aspiration and/or core needle biopsy for subclassifying lymphoma. Arch Pathol Lab Med. 2015; 139:245-51. DOI
  78. Hu Q, Naushad H, Xie Q. Needle-core biopsy in the pathologic diagnosis of malignant lymphoma showing high reproducibility among pathologists. Am J Clin Pathol. 2013; 140:238-47. DOI
  79. Amador-Ortiz C, Chen L, Hassan A. Combined core needle biopsy and fine-needle aspiration with ancillary studies correlate highly with traditional techniques in the diagnosis of nodal-based lymphoma. Am J Clin Pathol. 2011; 135:516-24. DOI
  80. Zelenetz AD, Abramson JS, Advani RH. NCCN clinical practice guidelines in oncology for non-Hodgkin’s lymphomas. J Natl Compr Canc Netw. 2010; 8:288-334. DOI
  81. Johl A, Lengfelder E, Hiddemann W. Core needle biopsies and surgical excision biopsies in the diagnosis of lymphoma-experience at the Lymph Node Registry Kiel. Ann Hematol. 2016; 95:1281-6. DOI

Affiliations

$authorString->getOrcid() =>

$authorString->getFullName() => Stefano Fratoni

$authorString->getUrl() =>

Stefano Fratoni

Department of Anatomic Pathology, St. Eugenio Hospital of Rome, Rome, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Magda Zanelli

$authorString->getUrl() =>

Magda Zanelli

Pathology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Maurizio Zizzo

$authorString->getUrl() =>

Maurizio Zizzo

Surgical Oncology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy; Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Francesca Sanguedolce

$authorString->getUrl() =>

Francesca Sanguedolce

Pathology Unit, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Foggia, Foggia, Italy;
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Valentina Aimola

$authorString->getUrl() =>

Valentina Aimola

Pathology Unit, University of Cagliari, Cagliari, Italy;
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Giulia Cerrone

$authorString->getUrl() =>

Giulia Cerrone

Pathology Unit, University of Cagliari, Cagliari, Italy;
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Linda Ricci

$authorString->getUrl() =>

Linda Ricci

Pathology Unit, University of Siena, Siena, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Alessandra Filosa

$authorString->getUrl() =>

Alessandra Filosa

Pathology Unit, Ospedale di Ascoli Piceno, Ascoli, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Giovanni Martino

$authorString->getUrl() =>

Giovanni Martino

Hematology Unit, CREO, Azienda Ospedaliera di Perugia, University of Perugia, Perugia, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Antonella Maria Fara

$authorString->getUrl() =>

Antonella Maria Fara

Pathology Unit, Department of Medical, Surgical and Experimental Surgery, University of Sassari, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Valerio Annessi

$authorString->getUrl() =>

Valerio Annessi

General Surgery Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Guastalla, Reggio Emilia, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Alessandra Soriano

$authorString->getUrl() =>

Alessandra Soriano

Gastroenterology Unit, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Stefano Ascani

$authorString->getUrl() =>

Stefano Ascani

Pathology Unit, Azienda Ospedaliera Santa Maria Terni, University of Perugia, Terni, Italy
non esiste orcidID ""

Copyright

© Società Italiana di Anatomia Patologica e Citopatologia Diagnostica, Divisione Italiana della International Academy of Pathology , 2020

How to Cite

[1]
Fratoni, S., Zanelli, M., Zizzo, M., Sanguedolce, F., Aimola, V., Cerrone, G., Ricci, L., Filosa, A., Martino, G., Fara, A.M., Annessi, V., Soriano, A. and Ascani, S. 2020. The broad landscape of follicular lymphoma: Part II. Pathologica - Journal of the Italian Society of Anatomic Pathology and Diagnostic Cytopathology. 112, 2 (Mar. 2020), 79-92. DOI:https://doi.org/10.32074/1591-951X-6-20.
  • Abstract viewed - 3383 times
  • PDF downloaded - 1625 times