If you don't remember your password, you can reset it by entering your email address and clicking the Reset Password button. You will then receive an email that contains a secure link for resetting your password
If the address matches a valid account an email will be sent to __email__ with instructions for resetting your password
Department of Pathology and Laboratory Medicine, Loyola University Medical Center, Maywood, IL, USADepartment of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, IL, USA
Address for correspondence: A/Prof Andres E. Quesada, The University of Texas MD Anderson Cancer Center, Department of Hematopathology, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
Address for correspondence: A/Prof Beenu Thakral, The University of Texas MD Anderson Cancer Center, Department of Hematopathology, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
T-cell neoplasms are a highly heterogeneous group of leukaemias and lymphomas that represent 10–15% of all lymphoid neoplasms. Traditionally, our understanding of T-cell leukaemias and lymphomas has lagged behind that of B-cell neoplasms, in part due to their rarity. However, recent advances in our understanding of T-cell differentiation, based on gene expression and mutation profiling and other high throughput methods, have better elucidated the pathogenetic mechanisms of T-cell leukaemias and lymphomas. In this review, we provide an overview of many of the molecular abnormalities that occur in various types of T-cell leukaemia and lymphoma. Much of this knowledge has been used to refine diagnostic criteria that has been included in the fifth edition of the World Health Organization. This knowledge is also being used to improve prognostication and identify novel therapeutic targets, and we expect this progress will continue, eventually resulting in improved outcomes for patients with T-cell leukaemias and lymphomas.
Although significant advances have been made in understanding the genetic underpinnings of T-cell malignances, they remain more complex and less well understood than their B-cell counterparts. In part, this disparity in understanding can be attributed to the presence of characteristic chromosomal translocations in some B-cell neoplasms that were recognised initially by using conventional cytogenetic analysis. For instance, 80–90% of follicular lymphomas carry t(14;18)(q32;q21)/IGH::BCL2 and >95% of mantle cell lymphoma cases carry t(11;14)(q13;q32)/CCND1::IGH. With the exception of one type of anaplastic large cell lymphoma and T-cell prolymphocytic leukaemia, most T-cell neoplasms do not carry non-cryptic characteristic translocations at such high frequencies. In part, the understanding of many T-cell neoplasms required the development of newer high-throughput technologies.
In this review, we provide an overview of the genetics of T-cell neoplasms stratified into two broad categories: immature and mature. We further subdivide mature T-cell neoplasms into four groups: (1) virally driven tumours; (2) those neoplasms with a predominantly leukaemic presentation; (3) tumours that present typically in lymph nodes; and (4) neoplasms that typically present at extra-nodal sites. Admittedly, this breakdown is imperfect as some mature T-cell neoplasms can fit into more than one category, but these categories are helpful in providing a structure for discussing the entities.
Immature T-cell neoplasms
Acute lymphoblastic leukaemia/lymphoma (ALL/LBL) is broadly classified into neoplasms of T-cell or B-cell lineage with rare NK-cell neoplasms. In the most recent version of the WHO classification of haematopoietic neoplasms, B-cell ALL/LBL (B-ALL/LBL) have nine different genetically defined groups whereas there is currently only one category of T-cell ALL/LBL.
However, it seems likely that T-ALL/LBL will be subdivided in the future as knowledge of pathogenesis and the development of biomarkers continues to expand.
T-lymphoblastic leukaemia/lymphoma
T-lymphoblastic leukaemia/lymphoma (T-ALL/LBL) is a neoplasm derived from immature blasts of the T-cell lineage.
The current 2017 WHO classification lists only one subtype of T-ALL/LBL: early T-cell precursor lymphoblastic leukaemia (ETP-ALL). T-ALL/LBL typically occurs in adolescents and young adults, but also can occur in the very young and the elderly.
T-ALL/LBL may present either as a lymphoma, often with a rapidly growing mediastinal mass and concomitant pleural effusions, or as leukaemia in the blood and/or bone marrow.
The most common genetic alterations in T-ALL/LBL are NOTCH1 activating mutations (50–70% of cases) and rarely t(7;9)(q34;q34) TRB::NOTCH1 fusions (<1%), a fusion with the gene encoding T-cell receptor beta chain that also results in activated NOTCH1.
Physiologically, NOTCH1 signalling is normally required to induce maturation of immature lymphoid precursor cells to become T-cells, and through NOTCH1 activation MYC is activated.
MYC is a well-established pro-growth signal that functions as a protooncogene in numerous malignancies. Unregulated NOTCH1 over expression through NOTCH1 activating alterations or loss of NOTCH1/MYC repression via FBXW7 alterations is thought to be one of the most typical routes of T-ALL/LBL pathogenesis.
In addition to NOTCH1 activation, activated TAL1 is another common driver of oncogenesis seen in 25–35% of cases of T-ALL/LBL. TAL1 is sometimes activated by a fusion involving TAL1 but is more commonly activated through a cryptic interstitial deletion of chromosome 1p32.
When TAL1 is activated through fusion, known partners include the genes that encode T-cell receptor alpha and T-cell receptor beta, TRA in t(1;14) and TRB in t(1;7), respectively. TAL1 is a transcription factor that is critical for normal erythropoiesis, but it is typically not expressed in other haematopoietic cells including normal immature lymphocytes/thymocytes and mature T-cells.
Alterations in TAL1 are mutually exclusive with alterations in TLX1, TLX3, HOXA9, HOXA10, NKX2-1, NKX2-2, LMO1, and LMO2, which are all mutually exclusive with one another.
Like TAL1, the transcription factors TLX1, TLX3, HOXA9, and HOXA10 also can be over expressed in immature T-cells where they can act as oncogenic signals. Often their over expression is the result of fusions or deletions, including fusions with TRB, TRG (the gene which encodes T-cell receptor gamma), or TRA.
Similarly, the transcription factor genes LMO1, LMO2, NKX2-1, and NKX2-2 can fuse with TRA or TRB resulting in aberrant expression in immature T-cells.
Associations between specific transcription factor expression and immunophenotype have been described, but to date there are no clinical recommendations to stratify therapy based on which transcription factor pathway is dysregulated.
All transcription factor aberrancies are typically seen in conjunction with NOTCH1/FBXW7 alterations, despite their mutual exclusivity with each other, suggesting both NOTCH1 signalling, and aberrant transcription factor over expression are needed for tumourigenesis in T-ALL/LBL.
In addition to up-regulation of the NOTCH1/FBXW7 pathway and transcription factor alterations, T-ALL/LBL has frequent deletions of CDKN2A/B in up to 70% of cases.
A proportion of T-ALL/LBL can have NUP214 fusions, deletions or mutations in PTEN and WT1, and loss of PHF6 in addition to many other genetic alterations.
Although many of these additional mutations are likely important to the pathogenesis in T-ALL/LBL cases, broadly most T-ALL/LBLs are driven by NOTCH1/FBXW7 activation, CDKN2A/B loss and overexpression of transcription factors that are not typically expressed in T-cells via their juxtaposition with T-cell receptor promoters.
Early T-cell precursor lymphoblastic leukaemia is the only WHO-defined subtype of T-ALL/LBL. As the name suggests, this entity is believed to arise from a T-cell committed immature precursor, but which has some features of an earlier uncommitted haematopoietic stem cell including myeloid and stem cell markers. Although ETP-ALL has both T-cell and myeloid differentiation by immunophenotype, the molecular alterations seen are far more consistent with myeloid malignances than typical T-ALL/LBL.
NOTCH1 alterations are less common, loss of CDKN2A/B is very uncommon, and transcription factor over expression, as occurs in many types of T-ALL/LBL, is not typical of ETP-ALL. Instead, ETP-ALL harbours mutations commonly seen in myeloid neoplasms, including alterations in FLT3, DNMT3A, NRAS, KRAS, IDH1 or IDH2, all of which are genetically rare in typical T-ALL/LBL.
BCL11B is another important transcription factor that has a role in differentiating precursor lymphocytes into T-cells. BCL11B is usually negative in ETP-ALL and is usually positive in most other cases of T-ALL/LBL.
B-cell lymphoma/leukaemia 11B (BCL11B) expression status helps distinguish early T-cell precursor acute lymphoblastic leukaemia/lymphoma (ETP-ALL/LBL) from other subtypes of T-cell ALL/LBL.
Mutations are somatic unless otherwise noted as germline.
, structural variants/fusion transcripts and copy number alterations) in T-lymphoblastic leukaemia/lymphoma
T-cell neoplasms (immature)
Genetic aberrations
References
T-lymphoblastic leukaemia/lymphoma
•
NOTCH1 activating mutations/alterations (50–70%); t(7;9)(q34; 934)TCRB::NOTCH1 fusions are rare (<1%)
•
FBXW7 loss of function mutations/alterations resulting in NOTCH1 overexpression (15–30%)
•
TAL1 (25–35%), TLX1, TLX3, HOXA9, HOXA10, NKX2-1, NKX2-2, LMO1, or LMO2 activating alterations/fusions (all alterations mutually exclusive of one another, seen in ∼53% and 66% of paediatric and adult T-ALL/LBL cases; typically seen in conjunction with NOTCH1/FBXW7 alteration)
B-cell lymphoma/leukaemia 11B (BCL11B) expression status helps distinguish early T-cell precursor acute lymphoblastic leukaemia/lymphoma (ETP-ALL/LBL) from other subtypes of T-cell ALL/LBL.
Most people acquire EBV infection in childhood and they experience either mild disease or are asymptomatic. When EBV infection is acquired in adolescence or adulthood, the result is often the clinical syndrome infectious mononucleosis.
As with many herpes viruses, after infection the host immune system is able to regain control over the virus but is not able to completely eliminate all viral elements.
Annually, EBV is the cause of nearly 200,000 malignancies world-wide, including neoplasms derived from B-cells, T-cells, NK-cells, and non-lymphoid cells, such as epithelial cells.
Many of these neoplasms are associated with inherited, acquired, or iatrogenic immune suppression but a number also occur in otherwise immunocompetent hosts.
Lymphomagenesis is driven by a set of EBV genes that are expressed even in latent cells. The viral proteins expressed promote tumour suppressor loss, for example, the viral protein EBNA1 promotes p53 degradation. EBV proteins also aberrantly activate oncogenic pathways, for example, LMP1 constitutively activates the JAK/STAT pathway.
Although lymphomagenesis is most well studied in relation to B-cell malignancies, EBV-driven T-cell malignancies likely have similar mechanisms of oncogenesis.
EBV-positive T-cell and NK-cell lymphoproliferative diseases of childhood
A disease category that includes four different entities: (1) systemic EBV-positive T-cell lymphoma of childhood; (2) chronic active EBV disease; (3) hydroa vacciniforme lymphoproliferative disorder; and (4) severe mosquito bite allergy.
Additionally, EBV-positive haemophagocytic lymphohistiocytosis, a benign disease which can induce major illness, can be discussed with this category of diseases.
EBV-positive haemophagocytic lymphohistiocytosis (EBV-positive HLH) is not a neoplasm per se, but rather an EBV-driven cytokine storm found in a small subset of children who have immune defects in EBV immunosurveillance.
A number of known inherited genetic mutations can lead to abnormal immune response and EBV-positive HLH (familial HLH), but some cases appear to be without such aberrations (non-familial).
PCR-based clonality assessment can show T-cell clones in EBV-positive HLH.
Systemic EBV+ T-cell lymphoma of childhood (sEBV-TNHL) is the most deadly of the aformentioned diseases but can be difficult to distinguish from benign EBV-positive HLH as both can be associated with haemophagocytic histiocytosis.
Although no specific chromosomal abnormality is seen in all, or even most cases of sEBV+TNHL, some commonalities are present: cases with alterations typically have a complex karyotype (three or more chromosomal alterations) and almost 40% of cases in the literature have add(9)(p24); additionally, alterations of chromosome 1, 7, 11, 17, 20, 21 and X were seen in over 20% of reported cases.
In this way, CAEBV is the bridge between the always non-neoplastic EBV-positive HLH and the always neoplastic and malignant sEBV+TNHL. CAEBV can be systemic (CAEBV of T and NK-cell type, systemic form; sCAEBV) or cutaneous as in hydroa vacciniforme lymphoproliferative disorder (HV-LPD) and severe mosquito bite allergy (SMBA). Few studies show CAEBV shares similar somatic mutations (e.g., DDX3X and KMT2D) alterations as seen in T-cell and NK-cell lymphomas indicating this as a premalignant condition.
sCAEBV patients have infectious mononucleosis symptoms (fever, fatigue, haepatosplenomegaly, skin rashes, and lymphadenopathy) that do not resolve after three months. A proportion of these cases have HLH, which may transform to the more deadly systemic EBV+ T-cell lymphoma. sCAEBV patients do typically produce IgG to EBV; however, this immunoglobulin response is unable to keep the EBV in check, and peripheral blood will find >102.5 copies/mg EBV DNA.
sCAEBV is characterised by T-cells or NK-cells expression of viral genes EBNA1, LMP1 and LMP2 with resulting downstream oncogenic pathways activated and tumour suppressors inhibited.
Chromosome alterations associated with CAEBV include deletion of 6q or gain of 6p in HV-LPD. Similarly, chromosome 6q losses are also seen in ENKTL suggesting commonalities in the molecular pathogenesis of these two diseases.
At present, these neoplasms are currently classified under the rubric peripheral T-cell lymphoma, not otherwise specified. However, data are abundant supporting this idea that these neoplasms will become a separate entity. Primary EBV positive nodal T-/NK-cell lymphoma shares an immunophenotype with ENKTL.
Although primary EBV-positive nodal T-/NK-cell lymphoma can extend extranodally, it typically does not involve the skin, gastrointestinal system or nose, unlike ENKTL.
One study of primary EBV-positive T/NK-cell lymphoma showed recurrent copy number aberrations in approximately 20% of cases, including loss of chr14q11.2 (100%), chr3q26.1 (67%), and chr22q11.23 (33%).
ENKTL is associated with EBV in virtually all cases which likely indicates a pathogenic role. The genetics of ENKTL are somewhat similar to some EBV-positive T-cell and NK-cell lymphoproliferative diseases of childhood, although ENKTL occurs almost exclusively in adults.
ENKTL is another disease driven by EBV infection which, intriguingly, can show a 30 base-pair deletion in the viral gene LMP1 that seems to be associated with an increased risk of lymphomagenesis.
The TSIM subtype is associated with JAK/STAT pathway activation, NK cell origin, TP53 mutation, genomic instability including deletion of 6q21 and amplifications of 9p24.1 and/or 17q21.2, as well as PD-L1/2 overexpression.
JAK/STAT activation can occur by a variety of mechanisms, including amplifications of chromosome 9p24 (JAK2) and 17q21 (STAT3, STAT5B), and mutations of STAT3, JAK3, and JAK1.
The HEA subtype is associated with more epigenetic changes through mutations in HDAC9, EP300 and ARID1A, NF-KB activation, T-cell origin, and T-cell receptor signalling pathway activation. Lastly, The MB subtype is associated with MYC over expression and a poor prognosis with a 3-year overall survival rate of approximately 38.5%. In contrast, the 3-year OS rate for the TSIM and HEA subtypes is 79.1% and 91.7%, respectively.
Cytogenetic information on ENKTL is limited and varied but many cases harbour chromosomal alterations, most commonly 6q deletions (50%), focal 1q gain (50%) and 17p losses (40%); the 6q21–q25 region of loss includes PRDM1, FOXO3, ATG5, AIM1, HACE1 and PTPRK and the loss of 17p11–p13 includes the TP53 gene.
HTLV-1 is a retrovirus that is transmitted most often from mother to child via breastfeeding, but also via sexual activity or blood transfusion. There are over 10 million people infected worldwide by HTLV-1.
Like another retrovirus, HIV, HTLV-1 primarily infects T-cells, but unlike HIV HTLV-1 is primarily asymptomatic and viral nucleic acids and proteins are not easily detected in blood.
ATLL is a T-cell neoplasm caused by HTLV-1 which can present clinically in four different ways: the acute leukaemic form, the lymphomatous variant, a chronic form, and a smoldering variant.
Fig. 1(A–D) Adult T-cell leukaemia/lymphoma. (A) Hematoxylin-eosin (H&E) stained section of lymph node shows effacement of the architecture by sheets of medium to large lymphoid cells with irregular nuclear membranes and a subset of large cells with single to multiple prominent nucleoli and moderate cytoplasm. Tumour cells are positive for (B) CD4, (C) CD25, and (D) FOXP3 in a patient known to be HTLV-1-positive in serum.
Recently, others using whole genome sequencing have shown that over 30% of ATLL cases have mutations in the gene CIC, most well known for its association with low grade gliomas.
CIC has two isoforms: a long form and a short form. In low grade gliomas, CIC mutations tend to be in the shared exons of both isoforms, but in ATLL, mutations occur in the exons of the long isoform only.
Copy number alterations or mutations of ATXN1 are also common. Physiologically ATXN1 and CIC interact to create a complex that represses transcription.
As 53% of ATLL cases have either ATXN1 or CIC alterations, it is likely that dysregulation of this complex is a critical driver of ATLL in addition to viral infection.
Mutations in CCR4 (C-C chemokine receptor 4) are also common, and most patients with ATLL show overexpression of CCR4 which is associated with skin involvement and worse prognosis. However, a study by Ishida et al. showed potential therapeutic application of mogamulizumab, a monoclonal antibody targeting CCR4.
Copy number alterations are not uncommon in ATLL. This includes 9p24 amplification leading to PDL1 amplification, seen in 10–20% of cases, which is more common in the aggressive subtype, but it is associated with poor prognosis independent of subtype. Other copy alterations include 9p21 deletion (CDKN2A, 20–30%), 13q32 deletion (20–30%), 6p22 deletion (ATXN1, 10–20%) and deletion of 6q21 (PRDM1, 10–20%).
JAK3 missense mutations most common p.A572, p.A573V (5–35%), STAT3 missense mutations most common p.D661Y (6–27%), and STAT5B missense mutations most common p.N642H (2–6%)
T-cell prolymphocytic leukaemia (T-PLL) is a rare but aggressive leukaemia that involves blood and bone marrow in virtually all patients, but can involve lymph nodes, skin, and other anatomical sites.
Recurring genetic rearrangements are strongly associated with this disease: most commonly an inversion of chromosome 14 which places the TRA locus adjacent to TCL1A or TCL1B resulting in oncogene TCL1A/B over expression (80% of cases).
In 10% of cases there is a translocation involving TRA on chromosome 14 with a homolog to TCL1A and TCL1B on the X chromosome, MTCP1, seen as t(X;14) by karyotype.
T-PLL usually has other chromosomal abnormalities especially loss of the short arm of chromosome 8, gain of the long arm of chromosome 8, or both as in isochromosome 8q (50–60%).
JAK/STAT signalling is altered in up to 75% of cases, most commonly via JAK3 activating mutations/alterations (21–34%) and STAT5B mutations (36%). JAK3 mutations have a prognostic significance in T-PLL.
T-cell large granular lymphocytic leukaemia (T-LGLL)
T-LGLL is a chronic indolent clonal proliferation of T-cell large granular lymphocytes involving the blood (2–20×109/L) and other organs which has persisted for more than six months without a clearly identifiable cause. The persistent lymphocytosis can be mild and reactive aetiologies must be excluded before a diagnosis of leukaemia is rendered.
It primarily affects patients 45–75 years of age who present with neutropenia and moderate splenomegaly. STAT3 mutations are fairly common, identified in 30–40% of cases.
STAT5B mutations appear more commonly in CD4-positive T-LGL associated with indolent disease, whereas this same mutation in CD8-positive T-LGL is associated with aggressive disease.
Gain-of-function mutations in STAT3 or STAT5B can be helpful in differentiating between reactive large granular lymphocytosis and T-LGLL. However, a subset of cases of T-LGLL are STAT3 and STAT5B wild-type and other recurring genetic or cytogenetic alterations have not yet been found in T-LGL.
T-cell large granular lymphocytic leukaemia (T-LGL)
• STAT3 (30–40%) or STAT5B (2–5%) activating mutations/alterations
• STAT5B mutations more common in CD4-positive T-LGL, associated with indolent disease whereas this mutation in CD8-positive T-LGL is associated with aggressive disease.
About 90% of patients with coeliac disease have inherited the HLA isoforms DQ2 or DQ8, which are thought to contribute directly to the development of gluten intolerance.
Patients with EATL typically have a preceding diagnosis of coeliac disease and EATL, often preceded by so-called ‘refractory coeliac disease’ where patients continue to have symptoms and histological crypt hyperplasia and villous atrophy with increased lymphocytes despite a gluten-free diet.
Refractory coeliac disease is stratified into two types: type I (polyclonal T-cell lymphocytosis) and type II (clonal T-cell lymphocytosis); type II refractory coeliac disease (RCD) is about 5-fold more likely to transform into EATL when compared with type I (Fig. 2).
Fig. 2(A–D) Enteropathy-associated T-cell lymphoma (EATL). (A) Tumour invades muscularis propria with ulceration of the overlying small intestinal mucosa. (B) The tumour infiltrate is composed of predominantly small to medium size lymphoid cells with admixed occasional large cells with irregular nuclear membranes and condensed chromatin, positive for (C) CD3 with a high proliferation index as shown by (D) Ki-67.
Type II RCD, on the other hand, demonstrates many genetic abnormalities as are seen in EATL; in one study, JAK1/STAT3 pathway mutations were seen in 80% of RCDII and 90% of EATL – with over 50% of all cases harbouring gain-of-function mutations in a single codon (1097) of JAK1.
Gains of chromosome 1q are seen in both RCD type II and EATL, with additional gains in chromosomes 5q, 9q (focal), and 7q and losses of chromosomes 8p, 9p, 13q, and 16q (focal) identified in EATL, but not in RCD type II.
A study by Nicolae et al. found that genes involved in the JAK/STAT pathway were mutated in approximately 50% of patients with EATL, involving mutations in STAT3 (∼20%), STAT5B (∼10%), JAK1 (∼20%), and JAK3 (∼10%).
Unlike EATL, MEITL is not associated with coeliac disease or DQ2/DQ8 HLA isoforms. Genetically, MEITL is also different from EATL. Although some chromosomal alterations, such as gains in 9q, are also seen in MEITL, the MYC locus (8q24) is much more likely to be amplified in MEITL than EATL, and chromosome 1q and 5q gains are much less common.
Mutations affecting the JAK/STAT pathway are common in MEITL, present in ∼80% of cases and involving STAT5B (∼60%), JAK3 (∼46%), SH2B3 (∼20%) and JAK1 in a small subset.
Mutations affecting the MAPK pathway are also common in MEITL and involve TP53 (∼33%), BRAF (∼26%), KRAS (∼20%) and NRAS (∼10%), which appear to be mutually exclusive.
Classically, patients present with skin patches that progress to plaques or tumour stage over the course of many years. The lymphomatous T-cells show epidermotropism and a proclivity for infiltrating the basal layer of the skin/basement membrane. MF has an ultra-violet light mutational signature, similar to epithelial and melanocytic skin cancers that arise as a result of unprotected sun exposure.
In a UV light signature, cytosine to thymine changes are seen throughout the genome and suggest that UV light may play a role in disease pathogenesis; however, UV therapy is sometimes used in the treatment of MF and the temporal relationship between UV therapy and molecular sequencing has been underexplored.
Although initially described as the leukaemic presentation or leukaemic transformation of MF, there is some evidence suggesting SS may be a distinct disease entity, at least in some cases.
A study by Wang et al. showed that Sézary syndrome is characterised by mutations and loss-of-function of TP53, as well as mutations in CCR4, PLCG1, FAS and in CARD11 in over 10% of cases. Homozygous loss of CDKN2A/B is also common (58% cases).
Others have described the genetics for mycosis fungoides and Sézary syndrome together, which adds complexity to the interpretation of the data. Larocca and Kupper described the following recurrent alterations in MF/SS: single nucleotide variants in MLL3, TP53, CARD11, ARID1A, STAT5B, ZEB1, FAS, PLCG1, and CDKN2A and copy number variants in TP53, ZEB1, STAT5B, ARID1A, CDKN2A, FAS, DNMT3A, ATM, PRKCQ, and TNFAIP3.
Genetic sequencing examining mycosis fungoides and Sézary syndrome separately, before and after UV light therapy is needed to better identify the genetic underpinnings and molecular biology of these diseases.
Adipocyte rimming by lymphoma cells is a key characteristic histological finding in SPTCL. Clinically, many patients have autoimmune disorders, especially systemic lupus erythematosus, and haemophagocytic syndrome may be a complication of SPTCL. In 2019, others reported that 11/13 studied patients with SPTCL had germline mutations in the gene HAVCR2.
HAVCR2 encodes TIM-3 a protein expressed on T-cells that regulates immune cell function; loss of TIM-3 is associated with SPTCL, immune over activation and HLH and provides a unifying molecular basis for the clinical features of this disease.
Recurring somatic mutations were not impressively identified in SPTCL, although there may be some increase in PI3K/AKT/mTOR pathway alterations (Li, Lu et al. 2018).
SPTCL with HAVCR2 mutations are more likely to have systemic illness including haemophagocytic lymphohistiocytosis, present at a younger age and have worse relapse-free survival than HAVCR2 wild-type SPTCL.
Two primary cutaneous CD30-positive T-cell lymphoproliferative disorders exist which are thought to be two ends of a spectrum of disease: (1) lymphomatoid papulosis (LyP), and (2) primary cutaneous anaplastic large cell lymphoma (C-ALCL, Fig. 3A–D). A third group of borderline lesions with features between LyP and C-ALCL also have been described.
LyP lesions are small, often multifocal, and clinically self-resolve but can, and often do, return. C-ALCL may follow LyP in some patients; it is a larger lesion (>2 cm), often solitary, that is less likely to resolve or regress. When these two lesions cannot be distinguished from each other the term borderline lesion may be used.
By definition, these neoplasms do not carry ALK fusions/rearrangements. DUSP22::IRF4 is seen in a small proportion of LyP cases and more frequently in C-ALCL (20–57%).
Less commonly, (4–15%) NPM1::TYK2 fusions can be seen in primary cutaneous CD30-positive T-cell lymphoproliferative disorders, and a small subset of C-ALCL have TP63 rearrangement.
Fig. 3(A–D) Primary cutaneous anaplastic large cell lymphoma. (A) A solitary, localised skin tumour/nodule on the cheek. (B) Diffuse dermal involvement by (C) medium to large anaplastic cells with round to oval irregular nuclei and abundant cytoplasm. (D) Tumour cells are positive for CD30 (membranous and golgi pattern).
Given their rarity, large studies examining the genetic landscape of each entity are limited. PCGD-TCL cases appear to have KRAS, NRAS, MAPK, MYC, MYCN, JAK3, STAT3 or STAT5B missense mutations as well as loss of ARID1A, CDKN2A, FBXW7, SOCS1, TP53 and FAS.
PC8AE-TCL cases have been described with multiple chromosomal gains and losses, suggestive of chromosomal instability, a complex karyotype and TP53 loss.
Other less common mutations found in HTCL include those involving chromatin modifying and DNA methylation genes such as INO80, ARID1B, TET2, TET3, SMARCA2, and DNMT3A.
Fig. 4(A–D) Hepatosplenic T-cell lymphoma. (A) H&E section of bone marrow core biopsy specimen shows hypercellular (80–90%) bone marrow with a subtle lymphoid infiltrate composed of (B) medium and occasional large lymphoid cells with irregular nuclear membranes and condensed chromatin. (C) Bone marrow aspirate smear shows lymphoma cells with inconspicuous nucleoli (D) CD3 immunohistochemical stain highlights lymphoma cells in prominent expanded intrasinusoidal pattern.
Whole exome sequencing reveals activating JAK1 and STAT3 mutations in breast implant-associated anaplastic large cell lymphoma anaplastic large cell lymphoma.
Targeted next generation sequencing of breast implant-associated anaplastic large cell lymphoma reveals mutations in JAK/STAT signalling pathway genes, TP53 and DNMT3A.
Next generation sequencing of breast implant-associated anaplastic large cell lymphomas reveals a novel STAT3-JAK2 fusion among other activating genetic alterations within the JAK-STAT pathway.
Whole exome sequencing reveals activating JAK1 and STAT3 mutations in breast implant-associated anaplastic large cell lymphoma anaplastic large cell lymphoma.
Next generation sequencing of breast implant-associated anaplastic large cell lymphomas reveals a novel STAT3-JAK2 fusion among other activating genetic alterations within the JAK-STAT pathway.
Anaplastic large cell lymphoma (ALCL) is a CD30-positive systemic T-cell lymphoma. Systemic ALCL primarily presents in lymph nodes but can also involve extranodal sites; the most common sites of non-nodal involvement are abdominal and thoracic organs, bone marrow, soft tissue, and skin, although systemic ALCL never affects the skin solitarily.
Most cases of systemic ALCL have characteristic morphology including hallmark cells and grow in a cohesive growth pattern with variable sinusoidal involvement. There are two types of systemic ALCL: anaplastic lymphoma kinase (ALK)-positive ALCL and ALK-negative ALCL.
ALK-positive ALCL carries chromosomal translocations that involve ALK which are most commonly detected by immunohistochemistry. The most common translocation is t(2;5)(p23;q35), NPM1::ALK seen in about 85% of ALK+ ALCL.
This translocation results in fusion of the N-terminus of NPM1 (5q35) containing an oligomerisation domain with the tyrosine kinase domain of ALK (2p23) resulting in constitutive activation of ALK tyrosine kinase activity. The second most common fusion, t(1;2)(q25;p23), TPM3::ALK, is seen in 13% of ALK+ ALCL. Remaining ALK translocation partners are seen in 1% of ALK + ALCL and include: ATIC on chromosome 2, TFG on chromosome 3, CLTC on chromosome 7, MSN on the X chromosome, TPM4 on chromosome 19, MYH9 on chromosome 22, and RNF213 on chromosome 17, among others.
These fusion proteins not only result in constitutive ALK tyrosine kinase activity but are also thought to promote a variety of other oncogenic mechanisms, including JAK/STAT pathway activation.
Anaplastic large cell lymphoma, ALK-negative (Fig. 5A–D)
Systemic ALCL that does not have an ALK alteration or ALK expression, is fittingly termed ALK-negative ALCL. Systemic ALK-negative ALCL clinically presents similarly to ALK-positive ALCL and can be morphologically indistinguishable. Patients have generalised lymphadenopathy, extranodal sites of disease, and possible bone marrow involvement; hallmark cells are typically identified.
One major clinical difference in presentation is age: ALK-negative ALCL is primarily a disease of middle-aged or older adults whereas ALK-positive ALCL is primarily diagnosed in children or young adults.
However, at least two distinct genetic subgroups have been identified. (1) DUSP22 rearrangements are seen in approximately 30% of systemic ALK negative ALCL cases.
Literature describing the prognosis of DUSP22 rearranged ALK negative ALCL is conflicting with some studies demonstrating a favourable outcome, similar to ALK-positive disease, and other studies suggesting a more aggressive course than ALK-positive disease.
DUSP22 rearrangement is associated with distinctive immunophenotype but not outcome in patients with systemic ALK-negative anaplastic large cell lymphoma.
(2) TP63 rearrangements occur in approximately 8% of ALK-negative ALCL cases and confer a very poor prognosis, with a 5-year overall survival of only 17%, worse than PTCL-NOS.
DUSP22 and TP63 rearrangements are almost entirely mutually exclusive and cases without ALK, TP63 and DUSP22 rearrangements are sometimes referred to as triple-negative ALCL.
The JAK/STAT pathway is also over activated in a subset of cases, with mutations in that pathway including JAK1 and STAT3 which may co-occur and synergise to promote oncogenesis and sustain neoplastic growth.
Whole exome sequencing reveals activating JAK1 and STAT3 mutations in breast implant-associated anaplastic large cell lymphoma anaplastic large cell lymphoma.
Although, the mechanism of over expression is unlike its sisters ERBB2 and EGFR, the ERBB4 over expression is not the result of duplication of the ERBB4 at the DNA level (Fig. 5).
Fig. 5(A–D) Anaplastic large cell lymphoma, ALK negative. (A) H&E of lymph node shows effaced architecture by cohesive sheets of large, pleomorphic neoplastic cells, a subset of which has eccentric, horseshoe-shaped/kidney shaped hallmark cells (white arrow). Mitotic figures are not infrequent in the background. Tumour cells are positive for (B) CD30 (membranous and golgi pattern) and negative for (C) ALK immunostain. (D) Fluorescence in situ hybridisation (FISH) dual colour break apart probe to detect DUSP22 rearrangement. Cells with red/green split signals are positive for rearrangement.
AITL is a lymphoma derived from T-follicular helper cells (TFH) with intermixed prominent blood vessels (high endothelial venules) and large B-immunoblasts. This neoplasm is designated as nodal TFH lymphoma of AITL type in the new WHO classification.
The B-cells in AITL can themselves progress to an EBV-positive B-cell lymphoma, but in most cases of AITL the B-cells are not lymphomatous despite their sometimes large size and T-cell clonality in most cases. In addition, 25–30% of AITL cases will have clonal B-cells identified by IGH clonality testing.
Interestingly, the genetic mutations seen in AITL most overlap with those seen in myeloid neoplasms: TET2 mutations are seen in 47–83% of AITL cases and DNMT3A alterations are seen in 26–38% of cases. These mutations are thought to occur in stem cells. IDH2 mutations are also seen in 20–45% of AITL
Integrative clinicopathological and molecular analyses of angioimmunoblastic T-cell lymphoma and other nodal lymphomas of follicular helper T-cell origin.
Integrative clinicopathological and molecular analyses of angioimmunoblastic T-cell lymphoma and other nodal lymphomas of follicular helper T-cell origin.
Integrative clinicopathological and molecular analyses of angioimmunoblastic T-cell lymphoma and other nodal lymphomas of follicular helper T-cell origin.
The genetic landscape of NTCL-TFH shows significant overlap with that of AITL, but as mentioned previously, IDH2 mutations appear less common in NTCL-NOS.
Rare cases of peripheral T-cell lymphoma derived from T-follicular helper cells selectively involve follicles. These neoplasms are known as follicular T-cell lymphoma (FTCL). FTCL does not have high endothelial venule growth or expanded follicular dendritic meshwork, as with AITL, and shows a follicular growth pattern instead of diffuse growth.
Peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS)
PTCL-NOS is a T-cell lymphoma which does not meet criteria for any of the more specific subtypes. This neoplasm is also the most common category of T-cell lymphoma, comprising 25–30% of all cases.
Although the molecular features of PTCL-NOS are heterogenous, recurring alterations have been identified which also appear different from other T-cell lymphomas, including AITL and anaplastic large cell lymphoma.
Gene expression profiling has identified at least two major subgroups of PTCL NOS: one with high expression of GATA3 (PTCL-GATA3), and another with high expression of TBX21 (PTCL-TBX1).
PTCL-GATA3 is associated with PI3K/mTOR pathway activation, a more aberrant genome, and an inferior prognosis (5-year overall survival of 19%). In contrast, PTCL-TBX21 is associated with NFKB pathway activation, a less aberrant genome and a 5-year overall survival of 38%.
Watatani et al. showed that TP53/CDKN2A alterations in PTCL-NOS were associated with marked chromosomal instability and a worse overall survival and were negatively correlated with TFH marker expression. Other recurring mutations include FAT1, PLCG1, CD28 and VAV1. Recurrent mutations seen in PTCL-TFH/AITL, including TET2, DNMT3A and RHOA, can also be seen in PTCL-NOS, but at far lower frequencies.
PTCL-NOS as a whole tends to show far greater genomic complexity than AITL.
Chromothripsis, a type of chromosomal instability with many chromosomal rearrangements in a small area, is seen in many PTCL-NOS cases by whole genome sequencing.
Furthermore, Maura et al. showed that co-occurrence of CDKN2A and PTEN deletions may be specific for PTCL-NOS, with absence of co-deletion in both ALCL and AITL.
PTCL-NOS cases can harbour a variety of structural variants or fusion transcripts including FYN::TRAF3IP2 fusions (∼17%), as were seen in nodal PTCL with TFH immunophenotype, as well as VAV1 rearrangements (∼11%).
Integrative clinicopathological and molecular analyses of angioimmunoblastic T-cell lymphoma and other nodal lymphomas of follicular helper T-cell origin.