Page 20 - 2020_07-Haematologica-web
P. 20

1762
Editorials
prior work, that the molecular pathogenesis of the CD4+ References
and CD8+ cases of ITLPD-GIT appears distinct.10,23 Thus, more formal separation of these phenotypic variants may be warranted in the future.
The current series presents both similarities with and differences from prior clinical reports.12 Endoscopic find- ings included multiple mucosal lesions, often with nodu- larity or polyps. Only one case was associated with mucosal ulceration. Most of the patients had a very pro- tracted clinical course, with two patients being alive 19 and 21 years after diagnosis. There is a small but signifi- cant risk of transformation, with disease progressing in two patients after 11 and 27 years of follow-up. A vari- ety of treatments were employed, with no patient stated to attain a complete remission.
In prior series, all patients had disease confined to the gastrointestinal tract, with extraintestinal dissemination seen only in patients with histological progression.9,10 However, Soderquist et al. report bone marrow involve- ment in three cases, all of which were detected prior to transformation. In one case bone marrow involvement was detected only through an unidentified cytogenetic abnormality; the bone marrow was morphologically nor- mal and lacked evidence of a monoclonal T-cell receptor gene rearrangement. This patient is alive with disease at 7 years after presentation, so the presence of bone mar- row involvement, if real, has had little clinical impact. Two additional patients were reported to have inguinal lymph node involvement, one of whom also had positive bone marrow. This latter case is the only patient classified as having Ann Arbor Stage IV disease. This 41-year old male was asymptomatic at presentation, and is untreated, being alive with disease at 1 year. Curiously, the remain- ing three patients said to have “biopsy-proven” involve- ment of lymph node or bone marrow were classified as stage IE at diagnosis. Presumably, the stated bone mar- row or lymph node involvement occurred at some later point during the clinical course. More data are needed to understand the clinical and biological significance of this extraintestinal dissemination, including molecular data to confirm involvement.
A remaining issue is the optimal therapy for ITLPD- GIT. Most of the data are anecdotal. A number of patients have been treated with a variety of chemotherapy regi- mens used in both B-cell and T-cell lymphomas.9 Most patients have failed to achieve any long-term benefit from conventional chemotherapy. The JAK-STAT path- way appears to be an attractive target, especially in patients with CD4+ disease, and in recent years there has been interest in the use of targeted agents for a variety of mature T-cell and NK-cell malignancies.24 Ruxolitinib is a JAK-inhibitor approved for use in myeloproliferative neo- plasms, and has shown some activity in cutaneous T-cell lymphomas with activation of the JAK-STAT pathway.24 Other agents under evaluation for T-cell and NK-cell lym- phomas include tofacitinib, pacritinib, and the histone deacetylase inhibitor, chidamide. The use of targeted agents in combination with either chemotherapy or immunotherapy may offer promise in the future.
1. Isaacson P, Wright D. Malignant histiocytosis of the intestine: its relationship to malabsorption and ulcerative jejeunitis. Hum Pathol. 1978;9(6):661-677.
2. Isaacson P, Spencer J, Connolly C, et al. Malignant histiocytosis of the intestine: a T-cell lymphoma. Lancet. 1985;2(8457):688-691.
3. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127:2375-90.
4. 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.
5. Egawa N, Fukayama M, Kawaguchi K, et al. Relapsing oral and colonic ulcers with monoclonal T-cell infiltration. A low grade mucosal T-lymphoproliferative disease of the digestive tract. Cancer. 1995;75(7):1728-1733.
6. Hirakawa K, Fuchigami T, Nakamura S, et al. Primary gastrointesti- nal T-cell lymphoma resembling multiple lymphomatous polyposis. Gastroenterology. 1996;111(3):778-782.
7. Ranheim EA, Jones C, Zehnder JL, Warnke R, Yuen A. Spontaneously relapsing clonal, mucosal cytotoxic T-cell lympho- proliferative disorder: case report and review of the literature. Am J Surg Pathol. 2000;24(2):296-301.
8. Isomoto H, Maeda T, Akashi T, et al. Multiple lymphomatous poly- posis of the colon originating from T-cells: a case report. Dig Liver Dis. 2004;36(3):218-221.
9. Perry AM, Warnke RA, Hu Q, et al. Indolent T-cell lymphoprolifera- tive disease of the gastrointestinal tract. Blood. 2013;122(22):3599- 3606.
10. MargolskeeE,JobanputraV,LewisSK,AlobeidB,GreenPH,Bhagat G. Indolent small intestinal CD4+ T-cell lymphoma is a distinct enti- ty with unique biologic and clinical features. PLoS One. 2013;8:e68343.
11. Perry AM, Bailey NG, Bonnett M, Jaffe ES, Chan WC. Disease pro- gression in a patient with indolent T-cell lymphoproliferative disease of the gastrointestinal tract. Int J Surg Pathol. 2019;27(1):102-107.
12. Soderquist CR, Patel N, Murty VV, et al. Genetic and phenotypic characterization of indolent T-cell lymphoproliferative disorders of the gastrointestinal tract. Haematologica. 2020;105(7):1895-1906.
13. Amador C, Greiner TC, Heavican TB, et al. Reproducing the molec- ular subclassification of peripheral T-cell lymphoma-NOS by immunohistochemistry. Blood. 2019;134(24):2159-2170.
14. Koskela HLM, Eldfors S, Ellonen P, et al. Somatic STAT3 mutations in large granular lymphocytic leukemia. N Engl J Med. 2012;366(20):1905-1913.
15. Nicolae A, Xi L, Pittaluga S, et al. Frequent STAT5B mutations in gd hepatosplenic T-cell lymphomas. Leukemia. 2014;28 (11):2244-2248. 16. Nicolae A, Xi L, Pham TH, et al. Mutations in the JAK/STAT and RAS signaling pathways are common in intestinal T-cell lymphomas.
Leukemia. 2016;30(11):2245-2247.
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. Chiarle R, Simmons WJ, Cai HY, et al. Stat3 is required for ALK- mediated lymphomagenesis and provides a possible therapeutic tar-
get. Nat Med. 2005;11(6):623-629.
19. 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.
20. Blombery P, Thompson E, Jones K, et al. Whole exome sequencing
reveals activating JAK1 and STAT3 mutations in breast-implant asso-
ciated anaplastic large cell lymphoma. 2016;101(9):e387-e390.
21. LaurentC,NicolaeA,LaurentC,etal.Genealterationsinepigenetic modifiers and JAK-STAT signaling are frequent in breast implant-
associated ALCL. Blood. 2020;135(5):360-370.
22. Xiao W, Gupta GK, Yao J, et al. Recurrent somatic JAK3 mutations
in NK-cell enteropathy. Blood. 2019;134(12):986-991.
23. Sharma A, Oishi N, Boddicker RL, et al. Recurrent STAT3-JAK2 fusions in indolent T-cell lymphoproliferative disorder of the gas-
trointestinal tract. Blood. 2018;131(20):2262-2266.
24. Shouse G, Nikolaenko L. Targeting the JAK/STAT pathway in T cell
lymphoproliferative disorders. Curr Hematol Malig Rep. 2019;14 (6):570-576.
haematologica | 2020; 105(7)


































































































   18   19   20   21   22