Vol 52, No 4 (2021)
Review article
Published online: 2021-08-31

open access

Page views 313
Article views/downloads 331
Get Citation

Connect on Social Media

Connect on Social Media

Treatment of peripheral T-cell lymphomas

Grzegorz Mazur1, Justyna Kanclerska1
Acta Haematol Pol 2021;52(4):334-339.

Abstract

Peripheral T-cell lymphomas (PTCLs) are rare neoplasms that recently have been the subject of much research into their complex pathophysiology. PTCLs are a heterogeneous group of tumors consisting of nodal and extranodal leukemic and cutaneous neoplasms. PTCLs are associated with complex biology and arduous pathology which is currently being studied. According to this research, the pathophysiology of PTCLs can be divided into intrinsic and extrinsic mechanisms. Among the intrinsic mechanisms, scientists have described JAK-STAT pathway deregulation, as well as different somatic mutations including RB1, PTEN, TP53 and structural changes to the receptors. Also, there are scientific papers that correlate Epstein-Bárr virus or human T-cell lymphotropic virus type 1 infections with the occurrence of the neoplasm. PTCLs are most likely to develop in Asian and African populations. Due to poor clinical outcomes, PTCL treatment is the subject of intense clinical research. As a result of that, new drugs have been approved by the Food and Drug Administration for use among patients with refractory PTCL: pralatrexate, an antifolate drug; romidepsin, belinostat, an inhibitor for histone deacetylase, and brentuximab vedotin, a CD30 antibody. Also, clinical trials with mogamulizumab are being carried out for PTCL treatment. In addition to this, lenalidomide, as a substance that regulates the immune system and has shown antineoplastic effect in several hematological studies, could possibly be considered as treatment.

Article available in PDF format

View PDF Download PDF file

References

  1. Vose J, Armitage J, Weisenburger D, et al. International T-Cell Lymphoma Project. International peripheral T-cell and natural killer/T-cell lymphoma study: pathology findings and clinical outcomes. J Clin Oncol. 2008; 26(25): 4124–4130.
  2. Anderson JR, Armitage JO, Weisenburger DD. Epidemiology of the non-Hodgkin's lymphomas: distributions of the major subtypes differ by geographic locations. Non-Hodgkin's Lymphoma Classification Project. Ann Oncol. 1998; 9(7): 717–720.
  3. Trempat P, Tabiasco J, Andre P, et al. Evidence for early infection of nonneoplastic natural killer cells by Epstein-Barr virus. J Virol. 2002; 76(21): 11139–11142.
  4. Inghirami G, Chan WC, Pileri S, et al. AIRC 5xMille consortium ‘Genetics-driven targeted management of lymphoid malignancies’. Peripheral T-cell and NK cell lymphoproliferative disorders: cell of origin, clinical and pathological implications. Immunol Rev. 2015; 263(1): 124–159.
  5. Weiss LM, Jaffe ES, Liu XF, et al. Detection and localization of Epstein-Barr viral genomes in angioimmunoblastic lymphadenopathy and angioimmunoblastic lymphadenopathy-like lymphoma. Blood. 1992; 79(7): 1789–1795.
  6. 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.
  7. Iqbal J, Wright G, Wang C, et al. Lymphoma Leukemia Molecular Profiling Project and the International Peripheral T-cell Lymphoma Project. Gene expression signatures delineate biological and prognostic subgroups in peripheral T-cell lymphoma. Blood. 2014; 123(19): 2915–2923.
  8. Shouse G, Nikolaenko L. Targeting the JAK/STAT pathway in T cell lymphoproliferative disorders. Curr Hematol Malig Rep. 2019; 14(6): 570–576.
  9. Gupta M, Maurer M, Stenson M, et al. In-vivo activation of STAT3 in angioimmunoblastic T cell lymphoma, PTCL not otherwise specified, and ALK negative anaplastic large cell lymphoma: implications for therapy. Blood. 2013; 122(21): 844–844.
  10. Dufva O, Kankainen M, Kelkka T, et al. Aggressive natural killer-cell leukemia mutational landscape and drug profiling highlight JAK-STAT signaling as therapeutic target. Nat Commun. 2018; 9(1): 1567.
  11. Fujiwara Si, Yamashita Y, Nakamura N, et al. High-resolution analysis of chromosome copy number alterations in angioimmunoblastic T-cell lymphoma and peripheral T-cell lymphoma, unspecified, with single nucleotide polymorphism-typing microarrays. Leukemia. 2008; 22(10): 1891–1898.
  12. Palomero T, Couronné L, Khiabanian H, et al. Recurrent mutations in epigenetic regulators, RHOA and FYN kinase in peripheral T cell lymphomas. Nat Genet. 2014; 46(2): 166–170.
  13. Feldman AL, Sun DX, Law ME, et al. Overexpression of Syk tyrosine kinase in peripheral T-cell lymphomas. Leukemia. 2008; 22(6): 1139–1143.
  14. Wilcox RA, Sun DX, Novak A, et al. Inhibition of Syk protein tyrosine kinase induces apoptosis and blocks proliferation in T-cell non-Hodgkin's lymphoma cell lines. Leukemia. 2010; 24(1): 229–232.
  15. Mahadevan D, Spier C, Della Croce K, et al. Transcript profiling in peripheral T-cell lymphoma, not otherwise specified, and diffuse large B-cell lymphoma identifies distinct tumor profile signatures. Mol Cancer Ther. 2005; 4(12): 1867–1879.
  16. Xiong J, Bian J, Wang L, et al. Dysregulated choline metabolism in T-cell lymphoma: role of choline kinase-α and therapeutic targeting. Blood Cancer J. 2015; 5: 287.
  17. Rohr J, Guo S, Huo J, et al. Recurrent activating mutations of CD28 in peripheral T-cell lymphomas. Leukemia. 2016; 30(5): 1062–1070.
  18. Kittipongdaja W, Wu X, Garner J, et al. Rapamycin suppresses tumor growth and alters the metabolic phenotype in T-cell lymphoma. J Invest Dermatol. 2015; 135(9): 2301–2308.
  19. Pechloff K, Holch J, Ferch U, et al. The fusion kinase ITK-SYK mimics a T cell receptor signal and drives oncogenesis in conditional mouse models of peripheral T cell lymphoma. J Exp Med. 2010; 207(5): 1031–1044.
  20. Lin ZX, Bai B, Cai QC, et al. High numbers of tumor-associated macrophages correlate with poor prognosis in patients with mature T- and natural killer cell lymphomas. Med Oncol. 2012; 29(5): 3522–3528.
  21. 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.
  22. Fujiwara SI, Yamashita Y, Nakamura N, et al. High-resolution analysis of chromosome copy number alterations in angioimmunoblastic T-cell lymphoma and peripheral T-cell lymphoma, unspecified, with single nucleotide polymorphism-typing microarrays. Leukemia. 2008; 22(10): 1891–1898.
  23. Zhao WL, Mourah S, Mounier N, et al. Vascular endothelial growth factor-A is expressed both on lymphoma cells and endothelial cells in angioimmunoblastic T-cell lymphoma and related to lymphoma progression. Lab Invest. 2004; 84(11): 1512–1519.
  24. Karwacz K, Bricogne C, MacDonald D, et al. PD-L1 co-stimulation contributes to ligand-induced T cell receptor down-modulation on CD8+ T cells. EMBO Mol Med. 2011; 3(10): 581–592.
  25. de Charette M, Houot R. Hide or defend, the two strategies of lymphoma immune evasion: potential implications for immunotherapy. Haematologica. 2018; 103(8): 1256–1268.
  26. Jain S, Van Scoyk A, Morgan EA, et al. Targeted inhibition of CD47-SIRPα requires Fc-FcγR interactions to maximize activity in T-cell lymphomas. Blood. 2019; 134(17): 1430–1440.
  27. Marzec M, Zhang Q, Goradia A, et al. Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, B7-H1). Proc Natl Acad Sci USA. 2008; 105(52): 20852–20857.
  28. Wilcox RA, Feldman AL, Wada DA, et al. B7-H1 (PD-L1, CD274) suppresses host immunity in T-cell lymphoproliferative disorders. Blood. 2009; 114(10): 2149–2158.
  29. Inghirami G, Chan WC, Pileri S, et al. AIRC 5xMille consortium ‘Genetics-driven targeted management of lymphoid malignancies’. Peripheral T-cell and NK cell lymphoproliferative disorders: cell of origin, clinical and pathological implications. Immunol Rev. 2015; 263(1): 124–159.
  30. Cheng J, Kydd AR, Nakase K, et al. Negative regulation of the SH2-homology containing protein-tyrosine phosphatase-1 (SHP-1) P2 promoter by the HTLV-1 Tax oncoprotein. Blood. 2007; 110(6): 2110–2120.
  31. d'Amore F, Gaulard P, Trümper L, et al. ESMO Guidelines Committee. Peripheral T-cell lymphomas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015; 26(Suppl 5): v108–v115.
  32. Foss F, Horwitz SM, Coiffier B, et al. Pralatrexate in patients with relapsed or refractory peripheral T-cell lymphoma: results from the pivotal PROPEL study. J Clin Oncol. 2011; 29(9): 1182–1189.
  33. Pro B, Advani R, Brice P, et al. National Comprehensive Cancer Network. Hodgkin lymphoma, version 2.2012 featured updates to the NCCN guidelines. J Natl Compr Canc Netw. 2012; 10(5): 589–597.
  34. Barta SK, Gong JZ, Porcu P. Brentuximab vedotin in the treatment of CD30+ PTCL. Blood. 2019; 134(26): 2339–2345.
  35. Domingo-Domènech E, Boumendil A, Climent F, et al. Lymphoma Working Party of the European Society for Blood and Marrow Transplantation, Lymphoma Working Party of the European Society for Blood and Marrow Transplantation. SGN-30: a basis for the effective treatment of CD30 positive hematopoietic malignancies. Expert Opin Investig Drugs. 2008; 17(12): 1883–1887.
  36. Moskowitz AJ, Ghione P, Jacobsen E, et al. Final results of a phase II biomarker-driven study of ruxolitinib in relapsed and refractory T-cell lymphoma. Blood. 2019; 134(Suppl 1): 4019–4019.
  37. Horwitz S, Feldman T, Hess B, et al. A phase 2 study of the dual SYK/JAK inhibitor cerdulatinib demonstrates good tolerability and clinical response in relapsed/refractory peripheral T-cell lymphoma and cutaneous T-cell lymphoma. Blood. 2019; 134(Suppl 1): 466–466.
  38. Hopfinger G, Nösslinger T, Lang A, et al. Lenalidomide in combination with vorinostat and dexamethasone for the treatment of relapsed/refractory peripheral T cell lymphoma (PTCL): report of a phase I/II trial. Ann Hematol. 2014; 93(3): 459–462.
  39. Lopez-Girona A, Mendy D, Ito T, et al. Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide. Leukemia. 2012; 26(11): 2326–2335.
  40. Lemonnier F, Safar V, Beldi-Ferchiou A, et al. Integrative analysis of a phase 2 trial combining lenalidomide with CHOP in angioimmunoblastic T-cell lymphoma. Blood Adv. 2021; 5(2): 539–548.
  41. Fujita Y, Nakaya A, Fujita S, et al. Mogamulizumab treatment of refractory peripheral T-cell lymphoma following autologous stem cell transplantation: A case report. Mol Clin Oncol. 2016; 4(2): 151–153.
  42. Ollila TA, Sahin I, Olszewski AJ. Mogamulizumab: a new tool for management of cutaneous T-cell lymphoma. Onco Targets Ther. 2019; 12: 1085–1094.
  43. Li X, Cheng Y, Zhang M, et al. Activity of pembrolizumab in relapsed/refractory NK/T-cell lymphoma. J Hematol Oncol. 2018; 11(1): 15.
  44. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res. 2020; 10(3): 727–742.
  45. Fiore D, Cappelli LV, Broccoli A, et al. Peripheral T cell lymphomas: from the bench to the clinic. Nat Rev Cancer. 2020; 20(6): 323–342.
  46. Grover NS, Savoldo B. Challenges of driving CD30-directed CAR-T cells to the clinic. BMC Cancer. 2019; 19(1): 203.
  47. Pinz K, Liu H, Golightly M, et al. Preclinical targeting of human T-cell malignancies using CD4-specific chimeric antigen receptor (CAR)-engineered T cells. Leukemia. 2016; 30(3): 701–707.