Context.—

In the 2017 revised World Health Organization classification of tumors of hematopoietic and lymphoid tissues, some mature T-cell lymphomas were reclassified and a few new provisional entities were established based on new data from clinical and laboratory studies. T follicular helper cell lymphoma is identified by T follicular helper cell markers. Anaplastic large cell lymphoma, ALK negative, is a better-defined entity based on genetic abnormalities, and breast implant–associated anaplastic large cell lymphoma is recognized as a provisional entity. The gastrointestinal T-cell lymphomas are reclassified, with addition of a new provisional entity, indolent T-cell lymphoproliferative disorder of the gastrointestinal tract, characterized by an indolent clinical course.

Objective.—

To review the diagnostic approaches to reclassified and newly established entities of mature T-cell lymphomas, focusing on significant immunophenotypic features and molecular genetic abnormalities. Relevant new discoveries after the publication of the 2017 World Health Organization classification are included.

Data Sources.—

Information from the literature most relevant to the 2017 World Health Organization revised classification and publications after 2016.

Conclusions.—

Incorporating clinical, morphologic, and immunophenotypic features usually provides sufficient evidence to reach a preliminary diagnosis of mature T-cell lymphoma. Molecular genetic studies can be very helpful for the final diagnosis and classification, especially in challenging cases. Some molecular genetic features have been found in breast implant–associated anaplastic large cell lymphoma, distinct from anaplastic large cell lymphoma, ALK negative. Immunohistochemical staining of 4 markers may enable further subtyping of peripheral T-cell lymphomas.

The accumulation of data from clinical and laboratory studies, especially molecular genetic studies, has led to reclassification of some mature T-cell lymphomas and establishment of a few new provisional entities in the 2017 revised World Health Organization (WHO) classification of tumors of hematopoietic and lymphoid tissues.1  Nodal T-cell lymphoma with T follicular helper cell (TFH) phenotype is reclassified. Anaplastic large cell lymphoma (ALCL), ALK negative, has been upgraded to an official entity, and better characterization of breast implant–associated ALCL enables it to become a separate provisional entity. The enteropathy-associated T-cell lymphoma types I and II are classified as separate entities, and indolent T-cell lymphoproliferative disorder of the gastrointestinal tract is established as a provisional entity based on its indolent clinical course. Gene expression profiling (GEP) studies have identified 2 subtypes of peripheral T-cell lymphomas (PTCLs), associated with the Th1 or Th2 differentiation pathway. Here we provide a brief review focusing on the diagnostic approaches and clinically significant molecular genetic abnormalities of these entities. Significant new discoveries after the publication of the 2017 WHO classification2  are included; the Epstein-Barr virus (EBV)–associated T-cell lymphoproliferative disorders in childhood and new entities/subtypes of primary cutaneous T-cell lymphoid neoplasms are not discussed in this review.

T follicular helper cell lymphoma (T-FHCL) is identified by the antigen markers expressed by the neoplastic cells, including CD10, BCL6, PD1, CXCL13, CXCR5, ICOS, and SAP. To define the TFH phenotype, expression of minimally 2 and ideally 3 or more of the above-listed markers is needed, in addition to CD4 and certain T-cell lineage–specific antigens.3 

The current WHO classification divides T-FHCL into 3 categories2 : angioimmunoblastic T-cell lymphoma (AITL), follicular T-cell lymphoma, and nodal PTCL with TFH phenotype. All 3 categories of T-FHCL have similar clinical manifestations.4,5  The distinction among the 3 categories is based on histopathologic features, including the distinction of morphologic variants within each category. Of note, the 3 categories of T-FHCL may have overlapping histopathologic features in concurrent biopsies of different lymph nodes or sequential biopsies in the same patients, and thus may represent morphologic variations of the same disease.5,6 Table 1 shows the diagnostic features distinguished among 3 categories of T-FHCL and their distinction from PTCL, not otherwise specified (NOS).

Table 1

Pathologic Features Distinguished Among 3 Categories of T Follicular Helper Cell Lymphoma and Peripheral T-Cell Lymphoma, Not Otherwise Specified (PTCL, NOS)

Pathologic Features Distinguished Among 3 Categories of T Follicular Helper Cell Lymphoma and Peripheral T-Cell Lymphoma, Not Otherwise Specified (PTCL, NOS)
Pathologic Features Distinguished Among 3 Categories of T Follicular Helper Cell Lymphoma and Peripheral T-Cell Lymphoma, Not Otherwise Specified (PTCL, NOS)

Angioimmunoblastic T-cell lymphoma is characterized by polymorphic cell infiltrate, increased high endothelial venules, and prominent follicular dendritic meshworks that can be highlighted by CD21/CD23 stains and are often expanded beyond lymphoid follicles; eosinophils are commonly increased, and neoplastic T cells are typically medium in size with clear cytoplasm and frequently form aggregates. Three morphologic variants, including perifollicular, interfollicular, and diffuse growth patterns, can be recognized7  (see Supplemental Figure S1 in the supplemental digital content containing 4 figures at https://meridian.allenpress.com/aplm in the August 2022 table of contents). The 3 patterns may represent chronologic manifestations or consecutive progression of the same disease, with perifollicular and interfollicular variants representing relatively early-phase AITL.8 

In follicular T-cell lymphoma, nodal architecture is completely or partially effaced by nodular (follicular) proliferation of medium-sized lymphocytes with a moderate amount of clear cytoplasm. Immunohistochemical analysis often shows an inverted pattern of B- and T-cell distribution, with T cells in the center of lymphoid follicles surrounded by B cells.5,6  CD21 often highlights well-formed follicular dendritic meshworks underneath the lymphoid follicles, as seen in follicular lymphoma. Two morphologic variants are recognized: follicular growth pattern and progressive transformation of germinal center (PTGC)–like growth pattern (see Supplemental Figure S2).5  The PTGC–like growth pattern, particularly when a low number of neoplastic T cells are present, is easy to miss without careful examination to confirm the morphologic atypia and phenotypic aberrancy of the neoplastic T cells. Cases with TFH phenotype that do not fit into either AITL or follicular T-cell lymphoma would be classified as nodal PTCL with TFH phenotype as a provisional entity.2 

All 3 categories of T-FHCL are frequently associated with latent EBV infection. This bystander infection could lead to clonal expansion of an EBV-positive B-cell population, resulting in EBV-positive B-cell lymphoproliferative disorder.9  Presence of Hodgkin/Reed-Sternberg–like cells, either EBV positive or EBV negative, frequently raises the differential diagnosis of classic Hodgkin lymphoma.10,11 

The diagnosis of T-FHCL often needs to be confirmed by demonstrating T-cell clonality. Ninety percent of AITLs have their T-cell clone detected by Biomed TCRG gene rearrangement and/or TCRB gene rearrangement analysis.12,13  Of note, in the perifollicular pattern of AITL and PTGC-like growth pattern of follicular T-cell lymphoma, T-cell clones may be missed because of low levels of neoplastic T cells. Angioimmunoblastic T-cell lymphoma often shares a signature mutation landscape with the other 2 categories of T-FHCL, including mutations in TET2, DNMT3A, and RHOA genes14,15 ; IDH2 mutation is relatively more frequent in AITL. This signature mutation profile can be used not only to confirm T-cell clonality, but also to distinguish from PTCL, NOS, which shows a more heterogeneous and different mutation landscape.16 TET2 and DNMT3A mutations are associated with advanced stage and short progression-free survival in AITL.14  In addition, specific mutation may become a useful marker for detecting minimal residual disease.

ALK-negative ALCL, defined as a CD30+ PTCL lacking expression of ALK protein but otherwise morphologically indistinguishable from ALCL, ALK positive, has been recognized as a distinct entity.2  Generally affecting older adults, with a median age of 58 years and the highest incidence in the sixth decade, ALK-negative ALCL is a genetically heterogeneous disease with widely disparate clinical outcomes. The 5-year overall survival varies significantly from 17% to 90% depending on the specific gene rearrangement status.17 

Most ALK-negative ALCLs show at least some suggestion of T-cell phenotype and partial to complete loss of one or more T-cell antigens, most commonly with absent expression of CD3, CD5, or TCR. Many cases express the cytotoxic markers TIA1, granzyme B, and/or perforin. Most cases show clonal rearrangement of TCR genes, whether or not they express T-cell antigens.17 

The 2017 revised WHO classification2  included the distinct gene rearrangements associated with a fraction of ALK-negative ALCLs. Approximately 30% of the cases harbor DUSP22 rearrangements and are associated with an up to 90% 5-year survival rate and favorable prognosis, similar to that seen in ALK-positive ALCL. Cases with DUSP22 rearrangements characteristically show a sheetlike growth pattern, “doughnut” cells by morphology, and less pleomorphism of tumor cells.18  The neoplastic cells typically lack cytotoxic marker expression.18  In addition, approximately 8% of the cases have TP63 gene rearrangements, which are associated with the worst prognosis and a 17% 5-year survival rate. These rearrangements are essentially mutually exclusive but rarely can occur concurrently.19  For prognostic stratification, it is recommended that all ALK-negative ALCLs undergo fluorescence in situ hybridization testing for rearrangements involving DUSP22 (DUSP22-IRF4 locus on 6p25.3) or TP63 (most commonly occurring as TBL1XR1/TP63 fusion [inv(3)(q26q28)]). The so-called triple-negative ALCL lacking rearrangements of ALK, DUSP22, and TP63 has an intermediate prognosis and 5-year survival rate of 42%.17 

Many GEP studies have been performed to elucidate ALK-negative ALCL biology, define the borders with other PTCL subtypes, and provide new genomic classifiers for stratification of patients. Agnelli et al20  found TNFRSF8, BATF3, and TMOD1 genes exhibited significantly higher expression in ALK-negative ALCL compared with PTCL, NOS. A 3-gene model was validated to separate ALK-negative ALCL from PTCL, NOS, with 97% accuracy.20  Cases with DUSP22 rearrangement lacked expression of genes associated with JAK1/STAT3 pathway activation but showed DNA hypomethylation, high expression of cancer-testis antigens, CD58 and HLA class II, and minimally expressed PD-L1.21  A microRNA expression profiling study22  demonstrated an 11-microRNA signature that distinguished ALK-negative ALCL from PTCL, NOS, with 90% probability. Inactivation of TP53 and/or PRDM123  and activating mutations of JAK1 and/or STAT3 genes are frequently seen in ALK-negative ALCL. Recurrent chimeras combining a transcription factor (NFkB2 or NCOR2) with a tyrosine kinase (ROS1 or TYK2) were also discovered in wild-type JAK1/STAT3 ALK-negative ALCL.24  Interestingly, constitutive STAT3 phosphorylation was observed in 43% of ALK-negative ALCLs, 76% of which had nonmutated JAK1/STAT3, suggesting alternative mechanisms of pathway activation.24  In addition, oncogenic truncated Erb-B2 receptor tyrosine kinase 4 (ERBB4) was reported to be ectopically expressed in 24% of ALK-negative ALCL, but not in ALK-positive ALCL or PTCL, NOS. Preliminary data suggest ERBB4 expression may be mutually exclusive with rearrangements of other genes such as TP63, DUSP22/IRF4, ROS, and TYK2, indicative of the presence of an ERBB4-positive subgroup.25  The diagnostic utility and clinical significance of these genomic abnormalities have to be further studied in a large panel of patients.

Breast implant–associated ALCL (BIA-ALCL) is a rare form of slow-growing PTCL that develops around implants used for breast augmentation or reconstruction. It is recognized as a provisional entity by the 2017 WHO classification2  with morphologic and immunophenotypic features indistinguishable from those of ALK-negative ALCL (see Supplemental Figure S3). All reported patients with BIA-ALCL to date are female, with a median age of 52 years (range, 28–87 years). Most patients present with a unilateral peri-implant effusion (seroma) and less frequently with a mass or hardening adjacent to the implant that involves breast parenchyma or soft tissue and/or regional lymph node. The median time from implant placement to diagnosis is 8 years, with a range of 1 to 30 years. Several studies reported a higher risk of disease development with textured implants compared with smooth, nontextured-surface implants.26  In most cases, explantation of the implant with a complete capsulectomy removing all disease, without chemotherapy, is considered to be curative and confers an excellent event-free and overall survival.27 

Fine-needle aspiration of peri-implant effusion can be submitted for cytology, cell block, flow cytometry study, and molecular testing. If a mass lesion is present, biopsy of the mass is the preferred diagnostic approach. Fixation and mapping of the capsulectomy specimen to select multiple representative sections are advised to assess for microscopic tumor involvement and capsular invasion.28,29  Based on the status of capsular involvement by tumor cells, the pathologic tumor stages are defined as T0 to T4.29  Tumors extending beyond the capsule and regional lymph nodes and/or incomplete capsulectomy are associated with worse prognosis.30 

Although these cases are rarely tested, the majority of tested cases show a complex karyotype and carry monoclonal TCRG and/or TCRB rearrangements. No ALK, DUSP22, or TP63 gene rearrangements are identified in BIA-ALCL.31,32  Recent studies32,33  have reported frequent activating mutations in the JAK1 and STAT3 pathways and loss of function alterations of epigenetic modifier. Gene expression profiling and RNA-seq studies provide further genetic justification to recognize BIA-ALCL as a separate disease entity.3436 

Enteropathy-associated T-cell lymphoma (EATL), formerly known as type I EATL, is strongly associated with celiac disease, occurs in populations of northern European origin, and shows polymorphic cellular composition, whereas monomorphic epitheliotropic intestinal T-cell lymphoma (MEITL), formerly known as type II EATL, shows no association with celiac disease, occurs predominantly in Asian and Hispanic populations, and is monomorphic in cytology.2  EATL and MEITL differ in terms of epidemiologic, clinical, histologic, immunophenotypic, and molecular genetic findings (see Table 2; Supplemental Figure S4).37,38  Transcriptome studies39  have demonstrated distinct expression signatures that separate EATL and MEITL, supporting the concept that the 2 lymphoma types are different entities.

Table 2

Differences of Clinical and Pathologic Features Between Enteropathy-Associated T-Cell Lymphoma (EATL) and Monomorphic Epitheliotropic Intestinal T-Cell Lymphoma (MEITL)

Differences of Clinical and Pathologic Features Between Enteropathy-Associated T-Cell Lymphoma (EATL) and Monomorphic Epitheliotropic Intestinal T-Cell Lymphoma (MEITL)
Differences of Clinical and Pathologic Features Between Enteropathy-Associated T-Cell Lymphoma (EATL) and Monomorphic Epitheliotropic Intestinal T-Cell Lymphoma (MEITL)

EATLs commonly lack surface CD3 and T-cell receptor expression.40  T-cell receptor αβ can be expressed in approximately 25% of EATL cases, whereas T-cell receptor γδ is seen only in rare cases.41  In contrast, the tumor cells in MEITL more commonly express T-cell receptor γδ.42  Immunophenotypic variation is not unusual in both lymphomas, and deviation in 1 or 2 markers is acceptable if other features are compatible. Of note, immunostaining of CD30 may be strong and uniform in the lymphoma cells in some EATL cases, making differentiation from ALCL challenging.43 

EATL and MEITL show some overlapping molecular genetic features. However, there are differences, including the gains of 1q and 5q more commonly found in EATL and the gains in 8q (MYC locus) more commonly found in MEITL. The mutational landscape is largely similar between EATL and MEITL. SETD2 is the most frequently mutated gene in EATL and MEITL, with different occurrence rates (approximately 15% versus up to 90%).44,45  In addition, the JAK-STAT signaling pathways are the most frequently mutated pathways, with frequent mutations in STAT5B as well as JAK1, JAK3, STAT3, SOCS1, GNAI2, SH2B3, and other genes.4446  Mutations in JAK3, STAT5B, and SH2B3 appear to be more frequent in MEITL compared with EATL. Furthermore, mutation in G protein alpha inhibiting activity polypeptide 2 (GNAI2) has been reported in up to 24% of MEITL cases, but not in EATL.46 

Indolent T-cell lymphoproliferative disorder of the gastrointestinal tract is a clonal T lymphoid cell proliferation infiltrating the lamina propria that can involve all gastrointestinal sites, more commonly small intestine and colon. The disease has an indolent clinical course, is persistent for years to decades, and lacks response to conventional chemotherapy. Disease progression and transformation are rare. The most appropriate management approach is watchful waiting.

Published cases show heterogeneous pathologic and molecular features.47,48  Generally, the disease is characterized by nondestructive expansion of small to medium-sized lymphoid cells with minimal cytologic atypia within the lamina propria of the mucosa. Focal infiltration in muscularis mucosae and submucosa can be seen. Immunophenotypically, the T cells express pan–T-cell markers (CD2, CD3, CD5, and CD7), but a subset of cases can show loss of CD5 or CD7. Most of the cases are CD4+ (approximately two-thirds) or CD8+ (nearly one-third). Rare cases show CD4 and CD8 dual positivity or negativity.47  CD56 is negative and T-cell receptor αβ is positive. The Ki-67 proliferation index is low (<5%–10%).38,41,47 

All cases show clonal T cell gene rearrangement, which is very helpful to support the diagnosis. Recurrent STAT3-JAK2 fusions have been reported in some CD4+ cases,47,49  whereas activating STAT3 mutations were not detected.48  Single-nucleotide polymorphism analyses have not demonstrated recurrent abnormalities, but rather some diverse chromosomal abnormalities.50 

Peripheral T-cell lymphoma, not otherwise specified (PTCL, NOS), is the most common mature T-cell lymphoma in North America and Europe, accounting for approximately one-third of all T-cell lymphoma cases. The category of PTCL, NOS, is considered a diagnostic wastebasket because it is a biologically and clinically heterogeneous disease and does not fit any specific WHO2  entities within T-cell lymphoproliferative disorders. Correspondingly, PTCL, NOS, demonstrates the most diverse morphologic and immunophenotypic characteristics with nodal and/or extranodal involvement. The lymphoma cells usually express CD3 and CD2 with down-regulation of CD5 and/or CD7. The majority of PTCL, NOS cases are CD4+/CD8, although cases with CD4/CD8+, CD4+/CD8+, and CD4/CD8 are sometimes seen. In addition to CD4, expression of a single TFH marker is allowed.

Gene expression profiling studies reveal that GATA3 expression is associated with inferior outcome of PTCL, NOS,51  and 2 mutually exclusive PTCL, NOS subgroups with significant difference in cell of origin and prognosis have recently been identified52 : one group demonstrates high expression of TBX21 (PTCL-TBX21), whereas the other group shows overexpression of GATA3 (PTCL-GATA3). TBX21 is a transcriptional master regulator of Th1 differentiation and GATA3 is a transcriptional master regulator of Th2 differentiation. The PTCL-TBX21 group constitutes approximately 50% PTCL, NOS, and shows Th1 phenotype with a gene signature enriched with interferon γ and NF-kB pathways. A subset of PTCL-TBX21 is positive for CD8 with overexpression of cytotoxic T-cell–related genes. The majority of PTCL-TBX21 cases show polymorphous appearance with abundant inflammatory cell infiltrates admixed with fewer tumor cells. The PTCL-GATA3 group accounts for approximately one-third of PTCL, NOS, and demonstrates a Th2 phenotype with a gene signature associated with MYC and PI3K pathways. Most PTCL-GATA3 cases comprise numerous large tumor cells with fewer background inflammatory cells. Prognostically, PTCL-TBX21 has a 5-year overall survival of 40%, whereas the 5-year overall survival of PTCL-GATA3 is close to 20%. Recently Amador et al53  proposed a simple immunohistochemical algorithm using 4 antibodies against TBX21, CXCR3, GATA3, and CCR4 to predict these 2 GEP-defined molecular subgroups (Figure). TBX21 and CXCR3 are for the PTCL-TBX21 subtype whereas GATA3 and CCR4 are for the PTCL-GATA3 subtype. Addition of CXCR3 and CCR4, chemokine receptors associated with the Th1 and Th2 phenotype, respectively, improved the accuracy of classification. This immunohistochemistry panel accurately reproduced the GEP results in the majority of cases (approximately 85%) in the original study, potentially providing a robust and practical tool in PTCL, NOS subclassification. Verification of this immunohistochemistry algorithm by a large study may be helpful to promote its application in practice and improve clinical decisions according to the underlying biology.

Immunohistochemical staining algorithm for subclassification of peripheral T-cell lymphoma, not otherwise specified (PTCL, NOS), based on cell of origin. The cutoff threshold of immunostaining positivity for TBX21 and CXCR3 is 20% and for GATA3 and CCR4 is 50%. The 4 markers can be stained simultaneously; however, the original study53  showed most cases can be classified by TBX21, CXCR3, and GATA3. Abbreviations: IHC, immunohistochemical staining; +, positive stain; −, negative stain.

Immunohistochemical staining algorithm for subclassification of peripheral T-cell lymphoma, not otherwise specified (PTCL, NOS), based on cell of origin. The cutoff threshold of immunostaining positivity for TBX21 and CXCR3 is 20% and for GATA3 and CCR4 is 50%. The 4 markers can be stained simultaneously; however, the original study53  showed most cases can be classified by TBX21, CXCR3, and GATA3. Abbreviations: IHC, immunohistochemical staining; +, positive stain; −, negative stain.

Close modal

Lennert lymphoma, currently named lymphoepithelioid lymphoma, was initially identified based on its morphologic characteristics. It is characterized by diffuse and sometimes interfollicular infiltrates composed of a variable number of neoplastic T cells admixed with a rich background of reactive epithelioid histiocytes. It has less extranodal involvement and portends a better prognosis compared with other PTCL, NOS. The WHO 2017 classification2  points out that the neoplastic cells are cytotoxic CD8+ T cells; however, the immunophenotypic features of the neoplastic cells is a subject of debate. Hartmann et al54  proposed that Lennert lymphoma cells have a nonactivated cytotoxic phenotype with TIA-1+ and granzyme B as well as a lack of TFH markers. However, Kurita et al55  reported that a significant subset of Lennert lymphomas comprise CD4+ neoplastic T cells with expression of one TFH marker. The TFH marker–positive Lennert lymphomas demonstrate a worse overall survival compared with TFH marker–negative counterparts despite the lack of a significant difference in histologic presentation. Limited analyses of genetic abnormalities and GEP also suggest enrichment of helper T cells in Lennert lymphoma.56 

Primary EBV+ nodal T/NK-cell lymphoma is recommended as a variant of PTCL, NOS, according to the WHO 2017 classification.2  Compared with EBV+ extranodal NK/T-cell lymphoma, this disease entity is associated with older age, advanced stage, and poorer survival. A recent copy number aberration study and GEP revealed that primary EBV+ nodal T/NK-cell lymphoma is different from the extranodal counterparts, with frequent loss of TCRA loci (on chromosome 14q11.2); upregulation of CD2, CD8, and PD-L1; and depressed expression of CD56.57  Therefore, it is crucial to include nodal involvement as one of the defining features for this entity.

The diagnosis and classification of mature T-cell neoplasms are frequently challenging. Integrating clinical, morphologic, and immunophenotypic features usually will provide evidence to reach a preliminary decision. Many cases need molecular genetic studies to finalize the diagnosis and refine the classification. There will be some cases for which experts may have difficulty determining or different opinions about the classification. Molecular genetic studies and mutation profiling can be very helpful for these challenging cases. Even when not required for the final diagnosis and classification, molecular and genomic tests may provide clinically significant information for management decisions.

1.
Hsi
ED.
2016 WHO classification update—what's new in lymphoid neoplasms
.
Int J Lab Hematol
.
2017
;
39
(suppl 1)
:
14
22
.
2.
Swerdlow
S,
Campo
E,
Harris
N,
et al, eds. World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues. Rev 4th ed
.
Lyon, France
:
International Agency for Research on Cancer;
2017
.
WHO Classification of Tumours; vol 2.
3.
Swerdlow
SH,
Campo
E,
Pileri
SA,
et al
The 2016 revision of the World Health Organization classification of lymphoid neoplasms
.
Blood
.
2016
;
127
(20)
:
2375
2390
.
4.
Mourad
N,
Mounier
N,
Briere
J,
et al
Clinical, biologic, and pathologic features in 157 patients with angioimmunoblastic T-cell lymphoma treated within the Groupe d'Etude des Lymphomes de l'Adulte (GELA) trials
.
Blood
.
2008
;
111
(9)
:
4463
4470
.
5.
Huang
Y,
Moreau
A,
Dupuis
J,
et al
Peripheral T-cell lymphomas with a follicular growth pattern are derived from follicular helper T cells (TFH) and may show overlapping features with angioimmunoblastic T-cell lymphomas
.
Am J Surg Pathol
.
2009
;
33
(5)
:
682
690
.
6.
Hu
S,
Young
KH,
Konoplev
SN,
Medeiros
LJ.
Follicular T-cell lymphoma: a member of an emerging family of follicular helper T-cell derived T-cell lymphomas
.
Hum Pathol
.
2012
;
43
(11)
:
1789
1798
.
7.
Attygalle
A,
Al-Jehani
R,
Diss
TC,
et al
Neoplastic T cells in angioimmunoblastic T-cell lymphoma express CD10
.
Blood
.
2002
;
99
(2)
:
627
633
.
8.
Rodriguez-Justo
M,
Attygalle
AD,
Munson
P,
Roncador
G,
Marafioti
T,
Piris
MA.
Angioimmunoblastic T-cell lymphoma with hyperplastic germinal centres: a neoplasia with origin in the outer zone of the germinal centre?: clinicopathological and immunohistochemical study of 10 cases with follicular T-cell markers
.
Mod Pathol
.
2009
;
22
(6)
:
753
761
.
9.
Attygalle
AD,
Kyriakou
C,
Dupuis
J,
et al
Histologic evolution of angioimmunoblastic T-cell lymphoma in consecutive biopsies: clinical correlation and insights into natural history and disease progression
.
Am J Surg Pathol
.
2007
;
31
(7)
:
1077
1088
.
10.
Moroch
J,
Copie-Bergman
C,
de Leval
L,
et al
Follicular peripheral T-cell lymphoma expands the spectrum of classical Hodgkin lymphoma mimics
.
Am J Surg Pathol
.
2012
;
36
(11)
:
1636
1646
.
11.
Nicolae
A,
Pittaluga
S,
Venkataraman
G,
et al
Peripheral T-cell lymphomas of follicular T-helper cell derivation with Hodgkin/Reed-Sternberg cells of B-cell lineage: both EBV-positive and EBV-negative variants exist
.
Am J Surg Pathol
.
2013
;
37
(6)
:
816
826
.
12.
van Krieken
JH,
Langerak
AW,
Macintyre
EA,
et al
Improved reliability of lymphoma diagnostics via PCR-based clonality testing: report of the BIOMED-2 Concerted Action BHM4-CT98-3936
.
Leukemia
.
2007
;
21
(2)
:
201
206
.
13.
Bruggemann
M,
White
H,
Gaulard
P,
et al
Powerful strategy for polymerase chain reaction-based clonality assessment in T-cell malignancies: report of the BIOMED-2 Concerted Action BHM4 CT98-3936
.
Leukemia
.
2007
;
21
(2)
:
215
221
.
14.
Yao
WQ,
Wu
F,
Zhang
W,
et al
Angioimmunoblastic T-cell lymphoma contains multiple clonal T-cell populations derived from a common TET2 mutant progenitor cell
.
J Pathol
.
2020
;
250
(3)
:
346
357
.
15.
Timmins
MA,
Wagner
SD,
Ahearne
MJ.
The new biology of PTCL-NOS and AITL: current status and future clinical impact
.
Br J Haematol
.
2020
;
189
(1)
:
54
66
.
16.
Watatani
Y,
Sato
Y,
Miyoshi
H,
et al
Molecular heterogeneity in peripheral T-cell lymphoma, not otherwise specified revealed by comprehensive genetic profiling
.
Leukemia
.
2019
;
33
(12)
:
2867
2883
.
17.
Parrilla Castellar
ER,
Jaffe
ES,
Said
JW,
et al
.
ALK-negative anaplastic large cell lymphoma is a genetically heterogeneous disease with widely disparate clinical outcomes
.
Blood
.
2014
;
124
(9)
:
1473
1480
.
18.
King
RL,
Dao
LN,
McPhail
ED,
et al
Morphologic features of ALK-negative anaplastic large cell lymphomas with DUSP22 rearrangements
.
Am J Surg Pathol
.
2016
;
40
(1)
:
36
43
.
19.
Karube
K,
Feldman
AL.
“Double-hit” of DUSP22 and TP63 rearrangements in anaplastic large cell lymphoma, ALK-negative
.
Blood
.
2020
;
135
(9)
:
700
.
20.
Agnelli
L,
Mereu
E,
Pellegrino
E,
et al
Identification of a 3-gene model as a powerful diagnostic tool for the recognition of ALK-negative anaplastic large-cell lymphoma
.
Blood
.
2012
;
120
(6)
:
1274
1281
.
21.
Luchtel
RA,
Dasari
S,
Oishi
N,
et al
Molecular profiling reveals immunogenic cues in anaplastic large cell lymphomas with DUSP22 rearrangements
.
Blood
.
2018
;
132
(13)
:
1386
1398
.
22.
Liu
C,
Iqbal
J,
Teruya-Feldstein
J,
et al
MicroRNA expression profiling identifies molecular signatures associated with anaplastic large cell lymphoma
.
Blood
.
2013
;
122
(12)
:
2083
2092
.
23.
Boi
M,
Rinaldi
A,
Kwee
I,
et al
PRDM1/BLIMP1 is commonly inactivated in anaplastic large T-cell lymphoma
.
Blood
.
2013
;
122
(15)
:
2683
2693
.
24.
Crescenzo
R,
Abate
F,
Lasorsa
E,
et al
Convergent mutations and kinase fusions lead to oncogenic STAT3 activation in anaplastic large cell lymphoma
.
Cancer Cell
.
2015
;
27
(4)
:
516
532
.
25.
Scarfò
I,
Pellegrino
E,
Mereu
E,
et al
Identification of a new subclass of ALK-negative ALCL expressing aberrant levels of ERBB4 transcripts
.
Blood
.
2016
;
127
(2)
:
221
232
.
26.
Doren
EL,
Miranda
RN,
Selber
JC,
et al
US epidemiology of breast implant-associated anaplastic large cell lymphoma
.
Plast Reconstr Surg
.
2017
;
139
(5)
:
1042
1050
.
27.
Miranda
RN,
Aladily
TN,
Prince
HM,
et al
Breast implant-associated anaplastic large-cell lymphoma: long-term follow-up of 60 patients
.
J Clin Oncol
.
2014
;
32
(2)
:
114
120
.
28.
Jaffe
ES,
Ashar
BS,
Clemens
MW,
et al
Best practices guideline for the pathologic diagnosis of breast implant-associated anaplastic large-cell lymphoma
.
J Clin Oncol
.
2020
;
38
(10)
:
1102
1111
.
29.
Lyapichev
KA,
Piña-Oviedo
S,
Medeiros
LJ,
et al
A proposal for pathologic processing of breast implant capsules in patients with suspected breast implant anaplastic large cell lymphoma
.
Mod Pathol
.
2020
;
33
(3)
:
367
379
.
30.
Quesada
AE,
Medeiros
LJ,
Clemens
MW,
Ferrufino-Schmidt
MC,
Pina-Oviedo
S,
Miranda
RN.
Breast implant-associated anaplastic large cell lymphoma: a review
.
Mod Pathol
.
2019
;
32
(2)
:
166
188
.
31.
Laurent
C,
Delas
A,
Gaulard
P,
et al
Breast implant-associated anaplastic large cell lymphoma: two distinct clinicopathological variants with different outcomes
.
Ann Oncol
.
2016
;
27
(2)
:
306
314
.
32.
Oishi
N,
Brody
GS,
Ketterling
RP,
et al
Genetic subtyping of breast implant-associated anaplastic large cell lymphoma
.
Blood
.
2018
;
132
(5)
:
544
547
.
33.
Laurent
C,
Nicolae
A,
Laurent
C,
et al
Gene alterations in epigenetic modifiers and JAK-STAT signaling are frequent in breast implant-associated ALCL
.
Blood
.
2020
;
135
(5)
:
360
370
.
34.
Di Napoli
A,
De Cecco
L,
Piccaluga
PP,
et al
Transcriptional analysis distinguishes breast implant-associated anaplastic large cell lymphoma from other peripheral T-cell lymphomas
.
Mod Pathol
.
2019
;
32
(2)
:
216
230
.
35.
Los-de Vries
GT,
de Boer
M,
van Dijk
E,
et al
Chromosome 20 loss is characteristic of breast implant-associated anaplastic large cell lymphoma
.
Blood
.
2020
;
136
(25)
:
2927
2932
.
36.
Oishi
N,
Hundal
T,
Phillips
JL,
et al
Molecular profiling reveals a hypoxia signature in breast implant-associated anaplastic large cell lymphoma
[published online
May
15,
2020]
.
Haematologica
.
37.
Swerdlow
SH,
Jaffe
ES,
Brousset
P,
et al
Cytotoxic T-cell and NK-cell lymphomas: current questions and controversies
.
Am J Surg Pathol
.
2014
;
38
(10)
:
e60
e71
.
38.
van Vliet
C,
Spagnolo
DV.
T- and NK-cell lymphoproliferative disorders of the gastrointestinal tract: review and update
.
Pathology
.
2020
;
52
(1)
:
128
141
.
39.
Mutzbauer
G,
Maurus
K,
Buszello
C,
et al
SYK expression in monomorphic epitheliotropic intestinal T-cell lymphoma
.
Mod Pathol
.
2018
;
31
(3)
:
505
516
.
40.
Tjon
JM,
Verbeek
WH,
Kooy-Winkelaar
YM,
et al
Defective synthesis or association of T-cell receptor chains underlies loss of surface T-cell receptor-CD3 expression in enteropathy-associated T-cell lymphoma
.
Blood
.
2008
;
112
(13)
:
5103
5110
.
41.
Soderquist
CR,
Bhagat
G.
Gastrointestinal T- and NK-cell lymphomas and indolent lymphoproliferative disorders
.
Semin Diagn Pathol
.
2020
;
37
(1)
:
11
23
.
42.
Chan
JK,
Chan
AC,
Cheuk
W,
et al
Type II enteropathy-associated T-cell lymphoma: a distinct aggressive lymphoma with frequent γδ T-cell receptor expression
.
Am J Surg Pathol
.
2011
;
35
(10)
:
1557
1569
.
43.
Arps
DP,
Smith
LB.
Classic versus type II enteropathy-associated T-cell lymphoma: diagnostic considerations
.
Arch Pathol Lab Med
.
2013
;
137
(9)
:
1227
1231
.
44.
Moffitt
AB,
Ondrejka
SL,
McKinney
M,
et al
Enteropathy-associated T cell lymphoma subtypes are characterized by loss of function of SETD2
.
J Exp Med
.
2017
;
214
(5)
:
1371
1386
.
45.
Roberti
A,
Dobay
MP,
Bisig
B,
et al
Type II enteropathy-associated T-cell lymphoma features a unique genomic profile with highly recurrent SETD2 alterations
.
Nat Commun
.
2016
;
7
:
12602
.
46.
Nairismagi
ML,
Tan
J,
Lim
JQ,
et al
JAK-STAT and G-protein-coupled receptor signaling pathways are frequently altered in epitheliotropic intestinal T-cell lymphoma
.
Leukemia
.
2016
;
30
(6)
:
1311
1319
.
47.
Montes-Moreno
S,
King
RL,
Oschlies
I,
et al
Update on lymphoproliferative disorders of the gastrointestinal tract: disease spectrum from indolent lymphoproliferations to aggressive lymphomas
.
Virchows Arch
.
2020
;
476
(5)
:
667
681
.
48.
Perry
AM,
Warnke
RA,
Hu
Q,
et al
Indolent T-cell lymphoproliferative disease of the gastrointestinal tract
.
Blood
.
2013
;
122
(22)
:
3599
3606
.
49.
Sharma
A,
Oishi
N,
Boddicker
RL,
et al
Recurrent STAT3-JAK2 fusions in indolent T-cell lymphoproliferative disorder of the gastrointestinal tract
.
Blood
.
2018
;
131
(20)
:
2262
2266
.
50.
Margolskee
E,
Jobanputra
V,
Lewis
SK,
Alobeid
B,
Green
PH,
Bhagat
G.
Indolent small intestinal CD4+ T-cell lymphoma is a distinct entity with unique biologic and clinical features
.
PLoS One
.
2013
;
8
(7)
:
e68343
.
51.
Wang
T,
Feldman
AL,
Wada
DA,
et al
GATA-3 expression identifies a high-risk subset of PTCL, NOS with distinct molecular and clinical features
.
Blood
.
2014
;
123
(19)
:
3007
3015
.
52.
Iqbal
J,
Wright
G,
Wang
C,
et al
Gene expression signatures delineate biological and prognostic subgroups in peripheral T-cell lymphoma
.
Blood
.
2014
;
123
(19)
:
2915
2923
.
53.
Amador
C,
Greiner
TC,
Heavican
TB,
et al
Reproducing the molecular subclassification of peripheral T-cell lymphoma-NOS by immunohistochemistry
.
Blood
.
2019
;
134
(24)
:
2159
2170
.
54.
Hartmann
S,
Agostinelli
C,
Klapper
W,
et al
Revising the historical collection of epithelioid cell-rich lymphomas of the Kiel Lymph Node Registry: what is Lennert's lymphoma nowadays?
Histopathology
.
2011
;
59
(6)
:
1173
1182
.
55.
Kurita
D,
Miyoshi
H,
Yoshida
N,
et al
A clinicopathologic study of Lennert lymphoma and possible prognostic factors: the importance of follicular helper T-cell markers and the association with angioimmunoblastic T-cell lymphoma
.
Am J Surg Pathol
.
2016
;
40
(9)
:
1249
1260
.
56.
Etebari
M,
Navari
M,
Agostinelli
C,
et al
Transcriptional analysis of Lennert lymphoma reveals a unique profile and identifies novel therapeutic targets
.
Front Genet
.
2019
;
10
:
780
.
57.
Ng
SB,
Chung
TH,
Kato
S,
et al
Epstein-Barr virus-associated primary nodal T/NK-cell lymphoma shows a distinct molecular signature and copy number changes
.
Haematologica
.
2018
;
103
(2)
:
278
287
.

Author notes

Supplemental digital content is available for this article at https://meridian.allenpress.com/aplm in the August 2022 table of contents.

X. Zhang and J. Zhou contributed equally to this review.

The authors have no relevant financial interest in the products or companies described in this article.

Presented, in part, at the Sixth Annual Chinese American Pathologists Association Diagnostic Course; October 10 and 11, 2020; virtual.

Supplementary data