Page 163 - 2020_07-Haematologica-web
P. 163

Genetic alterations in indolent GI T-cell LPD
from naïve T cells into bifunctional mucosal Th1/2 cells, similar to those described in primary immune responses against parasites, which help dampen inflammation,49 or derive from Th1 or Th2 cells that have undergone cytokine-mediated reprogramming to acquire a Th1/Th2 phenotype, with concomitant production of Th1 and Th2 cytokines.50 The phenotypic shift from a Th1/Th2 to Th2 profile over time, observed in one CD4+ case, suggests lin- eage (and possibly functional) plasticity of at least a sub- set of ITLPD. The majority of the CD8+ cases and the CD4-/CD8- ITLPD displayed a Tc2 phenotype.51 Besides orchestrating diverse functions in CD4+ T-helper cells, GATA3 also regulates the activation, homeostasis, and cytolytic activity of CD8+ T cells.52 The significance of T- bet/GATA3 co-expression in CD8+ ITLPD is unknown. It must be pointed out that despite the reported concor- dance between the transcriptional and protein expression profiles of T-bet and GATA3 in certain T-cell lymphomas,27 the definitive lineage (and function) of neo- plastic T cells cannot be ascertained based on the expres- sion of single lineage-associated transcription factors. Cytokine profiling and in vitro functional studies are
awaited for confirmation of our observations. Contrary to observations in peripheral T-cell lymphoma, not other- wise specified,27,53,54 however, an inferior prognostic impact of GATA3 expression was not apparent in our series of ITLPD.
In conclusion, our study reveals considerable immunophenotypic and genetic heterogeneity of GI ITLPD. We describe recurrent and novel genetic abnor- malities in different immunophenotypic subtypes of GI ITLPD which implicate deregulated cytokine signaling and epigenetic alterations in disease pathogenesis. It is hoped that future unbiased interrogation of ITLPD genomes and transcriptomes as well as mechanistic stud- ies will help to clarify the cell of origin and the functional consequences of the underlying genetic aberrations in these rare disorders, opening the door for targeted, less toxic and more effective therapies.
Acknowledgments
We would like to thank Raymond Yeh, PhD, for designing the primers, analyzing Sanger sequencing results, and generating electropherogram images of the IL2 rearrangements.
References
1. Swerdlow S, Campo E, Harris N, et al., edi- tors. World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon, France: IARC; 2016.
2. Foukas PG, de Leval L. Recent advances in intestinal lymphomas. Histopathology. 2015;66(1):112-136.
3. Wu XC, Andrews P, Chen VW, Groves FD. Incidence of extranodal non-Hodgkin lym- phomas among whites, blacks, and Asians/Pacific Islanders in the United States: anatomic site and histology differ- ences. Cancer Epidemiol. 2009;33(5):337- 346.
4. Delabie J, Holte H, Vose JM, et al. Enteropathy-associated T-cell lymphoma: clinical and histological findings from the International Peripheral T-Cell Lymphoma Project. Blood. 2011;118(1):148-156.
5. Tan SY, Chuang SS, Tang T, et al. Type II EATL (epitheliotropic intestinal T-cell lym- phoma): a neoplasm of intra-epithelial T- cells with predominant CD8αα phenotype. Leukemia. 2013;27(8):1688-1696.
6. Matnani R, Ganapathi KA, Lewis SK, Green PH, Alobeid B, Bhagat G. Indolent T- and NK-cell lymphoproliferative disorders of the gastrointestinal tract: a review and update. Hematol Oncol. 2017;35(1):3-16.
7. Xia D, Morgan EA, Berger D, Pinkus GS, Ferry JA, Zukerberg LR. NK-cell enteropa- thy and similar indolent lymphoprolifera- tive disorders. Am J Clin Pathol. 2018; 151(1):75-85.
8. Carbonnel F, D’Almagne H, Lavergne A, et al. The clinicopathological features of extensive small intestinal CD4 T cell infil- tration. Gut. 1999;45(5):662-667.
9. Carbonnel F, Lavergne A, Messing B, et al. Extensive small intestinal T-cell lymphoma of low-grade malignancy associated with a new chromosomal translocation. Cancer. 1994;73(4):1286-1291.
10. Hirakawa K, Fuchigami T, Nakamura S, et
al. Primary gastrointestinal T-cell lym- phoma resembling multiple lymphomatous polyposis. Gastroenterology. 1996;111(3): 778-782.
11. 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.
12. Perry AM, Warnke RA, Hu Q, et al. Indolent T-cell lymphoproliferative disease of the gastrointestinal tract. Blood. 2013;122(22):3599-3606.
13. Malamut G, Meresse B, Kaltenbach S, et al. Small intestinal CD4+ T-cell lymphoma is a heterogenous entity with common pathol- ogy features. Clin Gastroenterol Hepatol. 2014;12(4):599-608.
14. Edison N, Belhanes-Peled H, Eitan Y, et al. Indolent T-cell lymphoproliferative disease of the gastrointestinal tract after treatment with adalimumab in resistant Crohn’s coli- tis. Hum Pathol. 2016;57:45-50
15. 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.
naling pathways are frequently altered in epitheliotropic intestinal T-cell lymphoma. Leukemia. 2016;30(6):1311-1319.
20. Küçük C, Jiang B, Hu X, et al. Activating mutations of STAT5B and STAT3 in lym- phomas derived from gd-T or NK cells. Nat Commun. 2015;(6):6025.
21. Nicolae A, Xi L, Pham TH, et al. Mutations in the JAK/STAT and RAS signaling path- ways are common in intestinal T-cell lym- phomas. Leukemia. 2016;30(11):2245-2247.
22. van Dongen JJM, Langerak AW, Brüggemann M, et al. Design and standard- ization of PCR primers and protocols for detection of clonal immunoglobulin and T- cell receptor gene recombinations in sus- pect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4- CT98-3936. Leukemia. 2003;17(12):2257- 2317.
23. Margolskee E, Jobanputra V, Jain P, et al. Genetic landscape of T- and NK-cell post- transplant lymphoproliferative disorders. Oncotarget. 2016;7(25):37636-37648.
24. Trapnell C, Williams BA, Pertea G, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differen- tiation. Nat Biotechnol. 2010;28(5):511-
16. Sena Teixeira Mendes L, Attygalle AD, 515.
Cunningham D, et al. CD4-positive small T-cell lymphoma of the intestine present- ing with severe bile-acid malabsorption: a supportive symptom control approach. Br J Haematol. 2014;167(2):265-269.
17. 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.
18. Moffitt AB, Ondrejka SL, McKinney M, et al. Enteropathy-associated T cell lym- phoma subtypes are characterized by loss of function of SETD2. J Exp Med. 2017;214(5):1371-1386.
19. Nairismägi ML, Tan J, Lim JQ, et al. JAK- STAT and G-protein-coupled receptor sig-
25. Newman AM, Bratman S V., Stehr H, et al. FACTERA: a practical method for the dis- covery of genomic rearrangements at breakpoint resolution. Bioinformatics. 2014;30(23):3390-3393.
26. Tang G, Sydney Sir Philip JK, Weinberg O, et al. Hematopoietic neoplasms with 9p24/JAK2 rearrangement: a multicenter study. Mod Pathol 2019;32(4):490-498.
27. Iqbal J, Wright G, Wang C, et al. Gene expression signatures delineate biologic and prognostic subgroups in peripheral T- cell lymphoma. Blood. 2014;123(19):2915- 2924.
28. Perry AM, Bailey NG, Bonnett M, Jaffe ES, Chan WC. Disease progression in a patient with indolent T-cell lymphoproliferative
haematologica | 2020; 105(7)
1905


































































































   161   162   163   164   165