12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Cyclophosphamide as a first-line therapy in LGL leukemia

      letter

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Large granular lymphocyte (LGL) leukemia is a T or NK clonal disorder characterized by the tissue invasion of marrow, spleen and liver. Clinical presentation is dominated by recurrent infections associated with neutropenia, anemia, splenomegaly and autoimmune diseases, particularly rheumatoid arthritis. 1, 2, 3, 4 Recently, STAT3 and STAT5 mutations have been detected in T-LGL and in NK-LGL leukemias. 5, 6, 7, 8 These somatic mutations, coupled to other intrinsic and extrinsic mechanisms, are likely to induce constitutive activation of the JAK/STAT pathway thus contributing to maintenance of leukemic LGL survival. 9 These findings strongly suggest a common specific pathogenic pattern in T-LGL and NK-LGL leukemias and provide justification for consideration of the same treatment options. Indications for treatment are severe or symptomatic neutropenia, symptomatic or transfusion-dependent anemia or associated autoimmune diseases requiring therapy. There is no standard treatment for patients with LGL leukemia. All the largest series published in the literature (collecting data on more than 40 patients) are retrospective. Data are very heterogeneous and treatment outcome per single agent is available for very few patients. Immunosuppressive therapy remains the foundation of treatment including single agents that is, methotrexate, oral cyclophosphamide or cyclosporine. On the basis of an initial study showing very good overall response rate (ORR) using methotrexate, this drug has remained the most recommended option in LGL leukemia. 10 Oral low dose cyclophosphamide was first used in pure red cell aplasia associated with LGL leukemia. 11, 12 In a French series, cyclophosphamide was shown to be also efficient in neutropenic patients and for those who failed methotrexate. 13 Those results suggested that cyclophosphamide used as first-line therapy could be an interesting alternative to methotrexate. In this letter, we describe the encouraging results of cyclophosphamide used in a series of 45 previously untreated LGL leukemia patients. Patients suffering from LGL leukemia and treated with cyclophosphamide as first-line therapy were included in this retrospective study. Patients were screened from the Italian, French and USA Penn State registries. Patients gave their informed consent for data collection. The diagnosis of LGL leukemia was based on a chronic LGL peripheral blood expansion (>0.5 × 109/l), usually lasting for more than 6 months. Criteria for T-LGL leukemia included expression of LGL surface markers compatible with a typical T-cell (commonly αβ+ or γδ+/CD3+/CD8+/CD57+ and/or CD16+) phenotype associated with clonal rearrangement of TCRγ gene using PCR or clonal Vβ expression using flow cytometry. Criteria for NK-LGL lymphocytosis/chronic NK-LGL leukemia included expression of LGL surface markers compatible with a NK-cell (commonly CD3−/CD8+/CD16+ and/or CD56+) phenotype with more than 0.75 × 109/l circulating cells. 14, 15 Response to treatment was determined periodically on blood cell count and only best response was taken into account. Hematological complete response (CR) was defined by a normal blood count (hemoglobin (Hb)>12g/dl, platelets>150 × 109/l, absolute neutrophil count (ANC)>1.5 × 109/l and lymphocytosis<4 × 109/l) and LGL peripheral count in a normal range (<0.3 × 109/l). Molecular CR was based on hematological CR associated with a negative PCR analysis for CD3+ cases. Hematologic partial response was defined as an improvement in blood count specified as follows: ANC increasing more than 50% and reaching more than 0.5 but less than 1.5 × 109/l; Hb level increasing more than 2 g/dl and transfusion independent without reaching 12 g/dl level. Treatment failure was defined as a progressive disease (worsening of cytopenia or organomegaly) or a stable disease (none of the later given criteria met). Some patients received cyclophosphamide because of symptoms not related to cytopenia. For those patients, response criteria included clinical symptom resolution. Patients who received prednisone, granulocyte colony-stimulating factor or erythropoiesis-stimulating agent before or at the same time of cyclophosphamide were included in this retrospective study. For the descriptive analysis, qualitative variables were described using numbers and percentage, whereas medians and extremes were used to describe quantitative analyses. Qualitative variables were compared using χ 2 or Fisher's test. A total of 45 patients treated with cyclophosphamide as a first-line therapy for LGL leukemia from 1989 to 2012 were retrospectively included in this series. Clinical characteristics are described in Table 1. Starting doses were as follows: 100 mg/day (n=36), 50 to 75 mg/day (n=8) taken orally and 1 g IV/month (n=1). Median time from diagnosis to treatment was 3 months (range, 0–55). Treatment was initiated because of severe isolated neutropenia (n=16, 36%), neutropenia and anemia (n=2, 4%), transfusion-dependent anemia (n=15, 33%) and thrombocytopenia (n=5, 11% including three cases of idiopathic thrombopenic purpura). Seven non-cytopenic patients (15%) were treated for disease-associated LGL leukemia: neuropathy (n=3), vasculitis (n=1) and constitutive symptoms (n=3). ORR was 71% (32/45): there were 21 CR (47%) including three molecular responses (mCR) and 11 (24%) partial responses. ORR was 72% versus 68% for T-LGL and NK-LGL subtype, and 72% versus 67% for neutropenic and anemic patients, respectively (P=not significant). Patients treated for symptoms related to LGL leukemia had a 94% ORR (6/7). Eighteen patients (40%) were initially co-treated with prednisone and no significant impact was found for the response to cyclophosphamide (P=0.31). As well as, concomitant granulocyte colony-stimulating factor administration with cyclophosphamide did not influence the time to response and the ORR. The median time to reach best response was 4 months (range 0.8–21) and patients were treated for a median time of 6.4 months (range 0.5–33). Evidence of clinico-biological improvement was systematically observed within the 4 months following treatment initiation. Therefore, we assume that cyclophosphamide should be given for at least 4 months before changing drug regimen. Median treatment duration was 8.5 months (range 3.4–33) for responders, except one patient who received several courses of cyclophosphamide for 7 years. With a median time follow-up of 35 months (range 3.8–277), four out of the 32 responders relapsed (13%): one of these patients responded again to a short course of cyclophosphamide and was maintained subsequently on cyclosporine decided thereafter. The three others were switched to either methotrexate or cyclosporine. Eight patients experienced grade 1–2 toxicities (18%). Only three patients (7%) stopped treatment: one because of worsened anemia and two because of febrile neutropenia not obviously related to hematological toxicity. One patient experienced temporary worsening of anemia without stopping therapy. We previously mentioned that cyclophosphamide induces a very good ORR in LGL leukemia in de novo or relapsing patients. 14 The ORR of 71% described in our series confirmed and emphasized what has been reported in the literature with a total of 25 responders out of 38 patients treated as first line with cyclophosphamide (66% ORR) (Table 2). 11, 12, 13, 17, 18 We show that cyclophosphamide compares favorably to methotrexate given as a first-line therapy. In 1993, Loughran et al. 10 reported a 60% ORR in a prospective series of 10 patients receiving methotrexate at a weekly dose of 10 mg/m2. These results were less encouraging in two larger series. One comes from the prospective ECOG study showing a 37% ORR in 56 patients and the other is retrospective from the French registry and reported a 44% ORR in 36 patients. 13, 19 Furthermore, molecular response is rarely obtained and the incidence of relapse following methotrexate is, at least in French experience, estimated at 67%. 13 Our series demonstrates that cyclophosphamide used as first-line therapy is effective in T/NK-LGL leukemia, in both neutropenic and anemic patients. ORR and response duration seem encouraging. We recommend only 9–12 months of treatment. It seems sufficient to induce durable remissions and to avoid the complication of myelodysplastic syndromes/acute myeloid leukemia, which although rare is dependent on cumulative dose and length of exposure. For responding patients, tapering dose to 50 mg per day would be a reasonable option. Although recognizing the limits of a retrospective study, we suggest that cyclophosphamide could be an interesting alternative to methotrexate as first-line therapy in LGL leukemia. A prospective study comparing cyclophosphamide to methotrexate as first-line therapy is currently ongoing in France.

          Related collections

          Most cited references17

          • Record: found
          • Abstract: found
          • Article: not found

          STAT3 mutations unify the pathogenesis of chronic lymphoproliferative disorders of NK cells and T-cell large granular lymphocyte leukemia.

          Chronic lymphoproliferative disorders of natural killer cells (CLPD-NKs) and T-cell large granular lymphocytic leukemias (T-LGLs) are clonal lymphoproliferations arising from either natural killer cells or cytotoxic T lymphocytes (CTLs). We have investigated for distribution and functional significance of mutations in 50 CLPD-NKs and 120 T-LGL patients by direct sequencing, allele-specific PCR, and microarray analysis. STAT3 gene mutations are present in both T and NK diseases: approximately one-third of patients with each type of disorder convey these mutations. Mutations were found in exons 21 and 20, encoding the Src homology 2 domain. Patients with mutations are characterized by symptomatic disease (75%), history of multiple treatments, and a specific pattern of STAT3 activation and gene deregulation, including increased expression of genes activated by STAT3. Many of these features are also found in patients with wild-type STAT3, indicating that other mechanisms of STAT3 activation can be operative in these chronic lymphoproliferative disorders. Treatment with STAT3 inhibitors, both in wild-type and mutant cases, resulted in accelerated apoptosis. STAT3 mutations are frequent in large granular lymphocytes suggesting a similar molecular dysregulation in malignant chronic expansions of NK and CTL origin. STAT3 mutations may distinguish truly malignant lymphoproliferations involving T and NK cells from reactive expansions.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Discovery of somatic STAT5b mutations in large granular lymphocytic leukemia.

            Large granular lymphocytic (LGL) leukemia is characterized by clonal expansion of cytotoxic T cells or natural killer cells. Recently, somatic mutations in the signal transducer and activator of transcription 3 (STAT3) gene were discovered in 28% to 40% of LGL leukemia patients. By exome and transcriptome sequencing of 2 STAT3 mutation-negative LGL leukemia patients, we identified a recurrent, somatic missense mutation (Y665F) in the Src-like homology 2 domain of the STAT5b gene. Targeted amplicon sequencing of 211 LGL leukemia patients revealed 2 additional patients with STAT5b mutations (N642H), resulting in a total frequency of 2% (4 of 211) of STAT5b mutations across all patients. The Y665F and N642H mutant constructs increased the transcriptional activity of STAT5 and tyrosine (Y694) phosphorylation, which was also observed in patient samples. The clinical course of the disease in patients with the N642H mutation was aggressive and fatal, clearly different from typical LGL leukemia with a relatively favorable outcome. This is the first time somatic STAT5 mutations are discovered in human cancer and further emphasizes the role of STAT family genes in the pathogenesis of LGL leukemia.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              How I treat LGL leukemia.

              Large granular lymphocyte (LGL) leukemia is characterized by a clonal expansion of either CD3(+) cytotoxic T or CD3(-) NK cells. Prominent clinical features of T-LGL leukemia include neutropenia, anemia and rheumatoid arthritis (RA). The terminal effector memory phenotype (CD3(+)/CD45RA(+)/CD62L(-)CD57(+)) of T-LGL suggests a pivotal chronic antigen-driven immune response. LGL survival is then promoted by platelet-derived growth factor and interleukin-15, resulting in global dysregulation of apoptosis and resistance to normal pathways of activation-induced cell death. These pathogenic features explain why treatment of T-LGL leukemia is based on immunosuppressive therapy. The majority of these patients eventually need treatment because of severe or symptomatic neutropenia, anemia, or RA. No standard therapy has been established because of the absence of large prospective trials. The authors use low-dose methotrexate initially for T-LGL leukemia patients with neutropenia and/or RA. We recommend either methotrexate or oral cyclophosphamide as initial therapy for anemia. If treatment is not successful, patients are switched to either the other agent or cyclosporine. The majority of patients experience an indolent clinical course. Deaths infrequently occur because of infections related to severe neutropenia. As there are no curative therapeutic modalities for T-LGL leukemia, new treatment options are needed.
                Bookmark

                Author and article information

                Journal
                Leukemia
                Leukemia
                Leukemia
                Nature Publishing Group
                0887-6924
                1476-5551
                May 2014
                27 November 2013
                20 December 2013
                : 28
                : 5
                : 1134-1136
                Affiliations
                [1 ]Clinical Hematology Department-Rennes University Hospital , Rennes, France
                [2 ]Penn State Hershey Cancer Institute, Pennsylvania State College of Medicine , Hershey, PA, USA
                [3 ]Padua University School of Medicine, Department of Medicine, Hematology & Clinical Immunology Branch , Padua, Italy
                [4 ]Clinique de Cesson Sévigné-Rennes , Rennes, France
                [5 ]Clinical Hematology Department, Hôpital Estaing , Clermont-Ferrand, France
                [6 ]Hematology-Immunology and cell Therapy Department, Rennes University Hospital , Rennes, France
                [7 ]INSERM, CIC 0203 , France
                [8 ]Rennes 1 University , Rennes, France
                Author notes
                Article
                leu2013359
                10.1038/leu.2013.359
                4017255
                24280867
                6ded0a1c-d529-4da0-b328-ce613c65b92c
                Copyright © 2014 Macmillan Publishers Limited

                This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/

                History
                Categories
                Letter to the Editor

                Oncology & Radiotherapy
                Oncology & Radiotherapy

                Comments

                Comment on this article