Large Granular Lymphocyte Leukemia: Case Report of Chronic Neutropenia and Rheumatoid Arthritis-like Symptoms in a Child

2001 ◽  
Vol 4 (1) ◽  
pp. 94-99 ◽  
Author(s):  
Carol A. Blanchong ◽  
Randal Olshefski ◽  
Samir Kahwash

Lymphoproliferative disorders of large granular lymphocytes (LGL) are heterogeneous, with a clinical/pathologic spectrum ranging from a benign polyclonal expansion to an aggressive clonal disease. Often these lymphoproliferative disorders are associated with autoimmune disease. The clonal form of the disorder, LGL leukemia, typically occurs in older adults with a median age of 55 years at diagnosis. Pediatric cases are referred to in review articles; however, no detailed reports of T-cell LGL leukemia in children exist. This report illustrates a case of a child who presented initially at age 2 and 1/2 years with psoriasis, juvenile rheumatoid arthritis-like symptoms, and neutropenia. Bone marrow examinations obtained throughout his course have demonstrated progressive hypercellularity with increased reticulin fibers and replacement of the normal marrow elements by lymphocytes, which were later identified as large granular lymphocytes. Further testing with immunophenotyping by flow cytometry and T-cell receptor gene rearrangement studies revealed a monoclonal proliferation of large granular lymphocytes and confirmed a diagnosis of LGL leukemia. Although rare, large granular lymphocyte leukemia should be included in the differential diagnosis of chronic neutropenia in children.

2016 ◽  
Vol 69 (11-12) ◽  
pp. 376-378 ◽  
Author(s):  
Vanja Zeremski ◽  
Aleksandar Savic ◽  
Vesna Cemerikic-Martinovic ◽  
Ivana Milosevic ◽  
Marina Dragicevic ◽  
...  

Case report. A 41-year-old man presented with anemia, lymphocytosis and splenomegaly. T-cell large granular lymphocyte leukemia was diagnosed based on lymphocytosis of T-cell large granular lymphocytes, characteristic immunophenotype (CD3+, CD8+, CD16+, CD57+) of the lymphocytes and clonally rearranged T-cell receptor genes. Therapy indication was transfusion-dependent anemia. Initial cyclosporine therapy and low-dose oral methotrexate failed to control anemia and lymphocytosis. Yet, a complete clinical and hematological response (without molecular remission) was achieved and sustained when cyclosporine was reintroduced into the therapy. Conclusion. Our case confirms that cyclosporine therapy is effective in treating T-cell large granular lymphocyte leukemia and suggests that indefinite treatment may not be needed to maintain the response.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5290-5290
Author(s):  
Matthew R. Kovacs ◽  
Cyrus C. Hsia ◽  
Alejandro Lazo-Langner ◽  
Michael J. Kovacs

Abstract Background. T-Cell Large Granular Lymphocyte Leukemia (T-LGL Leukemia) is an extremely rare lymphoproliferative disorder which can originate from either mature T cells (CD3+) or natural killer cells (CD3-). T-LGL leukemia is a slowly progressing disease, rarely presenting with an aggressive course and in general patients have prolonged survival. Treatment is commonly monotherapy with methotrexate (with or without prednisone), cyclosporine, or cyclophosphamide. Herein we review our experience with the management of T-LGL in the last 15 years. Patients and Methods. We conducted a retrospective cohort study of all patients diagnosed with T-LGL at our center between 2000 and 2015. Patients were included if they had a positive study for T-Cell Receptor gene rearrangement. Response was evaluated using the criteria of Loughran et al (Leukemia 2015). Data was analyzed using descriptive statistics. Results. We included 23 patients, (16 male). All of the patients were caucasian with the exception of two. The average age at diagnosis is 65.1 years. Patients were treated for symptomatic cytopenias or splenomegaly with transfusions and/or chemotherapy. Six patients did not need any treatment. Fourteen of the 23 patients were treated with oral methotrexate monotherapy. The methotrexate dose ranged from 5mg/week to 15mg/week orally. Seven patients had a complete response, 2 had a partial response, 3 patients failed treatment, 1 patient was not evaluable, and 1 was stable. Four patients were treated with methotrexate and prednisone. Two patients obtained a complete response, with 1 partial response, and 1 patient was not evaluable due to the short duration of the prescription. Nine patients were treated with oral cyclophosphamide at doses of 50mg or 100mg po daily. Four patients achieved a complete response, 2 patients achieved a partial response, while the other 3 patients did not respond. Only 2 patients were treated with oral cyclosporine, at 400mg/day and at 500mg/day. One patient failed treatment while the other is still on therapy. Two patients died while on the study at 22 and 42 months, both deaths were due to other illnesses not associated with T-LGL Leukemia. The median time since diagnosis for the remaining patients is 41 months. Conclusion. T-LGL is a rare lymphoproliferative disorder. Most patients responded to simple oral therapy with methotrexate or cyclophosphamide and patients generally have prolonged survivals. Disclosures Lazo-Langner: Bayer: Honoraria; Pfizer: Honoraria. Kovacs:Daiichi Sankyo Pharma: Research Funding; LEO Pharma: Honoraria; Bayer: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1513-1513
Author(s):  
Aaron D. Viny ◽  
Hideki Makashima ◽  
Jungwon Huh ◽  
Karl S. Theil ◽  
Lukasz Gondek ◽  
...  

Abstract Large granular lymphocyte leukemia (LGL leukemia) is a semiautonomous clonal lymphoproliferation of cytotoxic T cells associated with various immune cytopenias. Within the spectrum of acquired immune-mediated bone marrow failure states, LGL leukemia can serve as monoclonal model of usually polyclonal T cell-mediated pathology. Mechanisms of unopposed clonal expansion of LGL cells in many aspects resemble true lymphoma and are not well understood. In addition to its reactive character, intrinsic clonal defects may be present in some patient with LGL, in particular those with more pronounced lymphoproliferative features. However, unlike in B-cell lymphomas, recurrent chromosomal abnormalities have not been frequently identified in LGL leukemia using traditional metaphase karyotyping techniques. We have applied Affymetrix 250K single nucleotide polymorphism-arrays (SNP-A) and 6.0 SNP-A in 28 patients with LGL leukemia, to elicit a far higher resolution of chromosomal content through genomic mapping of individual SNP and respective copy number analysis in LGL leukemia. SNP-A based cytogenetics have been applied successfully to MDS patients and has increased prognostic reliability. Blood mononuclear cells containing high proportion of clonal cells as determined by TCR Vβ flow cytometry were used as a source of DNA. For comparison, a large number of control blood and marrow specimens (N=119 for 6.0 and 124 for 250K SNP arrays) were analyzed. Data were processed using Genotyping Console v2.1 software (Affymetrix, Santa Clara, CA). After exclusion of known copy number variants (CNV) referenced in public databases and our own set of 178 normal controls, we found distinct chromosomal changes in 16/28 (57%) of LGL leukemia patients. Consensus regions of deletion/gain or uniparental disomy (UPD) were identified. The most common abnormalities included either UPD or copy number loss of chromosome 3q21.2–q21.31, which was identified in 6 (21%) patients. This region harbors CD86, the gene encoding the B7.2 protein responsible for T cell activation and regulation through costimulatory mechanisms. Copy number loss/UPD was also identified at chromosome 1p31.1–p32.3 in 4 (18%) patients. Interestingly, copy number gain in this same region was identified in 2 (9%) patients suggesting that this region may correspond to either a new, infrequent germ line encoded CNV or represents a somatic microdeletion. Recent studies indicated a role for SIL/SCL, which resides at this locus, in V(D)J recombination, lymphocyte development, and maturation. Additional conserved chromosomal abnormalities included copy number gains in 14q (11%) and copy number loss at 11p15 (14%), all possibly representing germ line or somatic CNV physiologically acquired during lymphocyte ontogenesis. We compared chromosomal lesions identified in our LGL cohort with clinical features including age, presence of neutropenia, anemia, thrombocytopenia, splenomegaly, immunophenotype, and degree of clonal expansion. No difference in clinical course was found between patients with and without cytogenetic abnormalities with regard to type and severity of cytopenias or size of the LGL clone. However, patients with 1p31.1–p32.3 deletions were found to have lesser degree of neutropenia compared to patients without 1p31.1–p32.3 deletions (p<0.001). While these data may reflect chromosomal variants and random somatic abnormalities, the recurrent loss of heterozygosity and gains within several loci with known regulatory function for lymphocyte biology suggests the involvement of these defects in the mechanism of clonal evolution.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 6590-6590
Author(s):  
Kirsten Marie Boughan ◽  
Thomas P. Loughran

6590 Background: The purpose of this study is to analyze patients enrolled in the LGL leukemia registry to distinguish the similarities between LGL leukemia and rheumatoid arthritis in order to access overlapping immune mechanisms that may be responsible for neutrophil mediated destruction. Methods: A retrospective chart review was performed on 79 patients enrolled in the LGL registry at Penn State Cancer Institute. All patients enrolled in the study had a diagnosis of both rheumatoid arthritis and potentially LGL leukemia. Data was collected for age, sex, RF factor positivity, family history, autoimmune disease, T-cell receptor gene rearrangement, and bone marrow invasion. Results: Of 79 patients the mean age of onset for LGL leukemia was 60 years old with no discrepancy noted between sexes, 37 M, 42 F. 49 patients were positive for rheumatoid factor. 27 patients had rheumatoid arthritis in a first degree relative with no discrimination between maternal or paternal inheritance. 22 patients were positive for any other autoimmune process. 60 patients were positive for T-cell receptor gene rearrangement. Of the remaining 19 patients that were negative for T-cell receptor rearrangement, 12 had evidence of bone marrow invasion (CD3/CD8+ infiltrate in >20% bone marrow) and two showed bone marrow invasion of NK cell LGL (CD3/CD8-, CD57+) (Table). Conclusions: Patients with T cell LGL leukemia and rheumatoid arthritis appear to be clinically similar with regard to age, duration of disease, and other autoimmune disorders as patients with rheumatoid arthritis alone. Our patient population showed those with TLGL and RA also tends to have a positive family history of RA in up to 20% as opposed to 5-10% in RA patients. Given that RA and TLGL have a significantly higher expression of the HLA-DR4 haplotype than healthy patients, it is conceivable that with shared genetic alterations, and gene environment interactions that may promote posttranslational modification, there may be a loss of tolerance resulting in T cell activation, and eventual transformation into a T cell clone. [Table: see text]


1997 ◽  
Vol 15 (2) ◽  
pp. 81-91 ◽  
Author(s):  
Margarida Lima ◽  
Maria Dos Anjos Teixeira ◽  
Ana Helena Ribeiro Dos Santos ◽  
Maria Luís Queirós ◽  
Benvindo Justiça

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1094-1094
Author(s):  
Aleksandr Lazaryan ◽  
Nelli Bejanyan ◽  
Aaron D. Viny ◽  
Medhat Askar ◽  
Pearlie K Burnette ◽  
...  

Abstract Abstract 1094 Poster Board I-116 T cell large granular lymphocyte leukemia (T-LGL) is a rare lymphoproliferative disorder marked by clonal expansion of cytotoxic T lymphocytes (CTL). T-LGL may be a result of clonal outgrowth from initially polyclonal CTL responses seen in the context of viral infections, autoimmune conditions, or tumor surveillance. Similar to classic autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis or aplastic anemia, immunogenetic predisposition to T-LGL is suggested by association with certain immunogenetic polymorphisms including human leukocyte antigen (HLA) and killer immunoglobulin receptor (KIR). In addition to KIR-ligand/KIR interactions, the quality of CTL may be determined by the binding between the major histocompatibility complex class I chain-related gene A (MICA) and its ligand NKG2A. Over fifty five MICA alleles have been documented to date. A number of autoimmune and oncologic conditions such as systemic lupus erythematosus, rheumatoid arthritis, psoriasis, Crohn's disease, cervical cancer, or oral squamous cell carcinoma were all reported to be associated with MICA polymorphism. We hypothesized that specific MICA polymorphisms may be associated with exaggerated CTL responses in T-LGL and may therefore impact clinical features of the disease such as immune cytopenias. We have collected a large well annotated cohort of patients with T-LGL (n=86). HLA, MICA, and KIR alleles were resolved by established molecular techniques. Diagnosis of T-LGL was established by flow cytometry, T cell receptor γ chain rearrangement, Vβ typing, and assessment of peripheral blood smear. Categorical and survival methods of data analysis were used to examine the association between MICA, HLA, and KIR polymorphisms with type and degree of cytopenias, LGL T cell count, response to therapy, splenomegaly, and overall survival. Caucasians accounted for 96.5% of the study cohort (median age, 64 years; 55% males). Neutropenia, anemia, and thrombocytopenia were found in 63.4%, 50%, and 23.5% of the patients, respectively. Bicytopenia and pancytopenia were found in 26% and 13% of subjects, respectively. Median LGL T cell count was 1800 cells/μL (range, 280-20,580 cells/μL). Splenomegaly was found in 47% of patients. Compared to healthy controls (2N=308), our cohort was overrepresented by MICA*A5.1 (population frequency, 0.59 in T-LGL vs. 0.37 in controls, p<0.001). As opposed to the patients carrying MICA*A4 (p=0.01), those with MICA*A5.1 (p=0.025) or MICA*008 (p=0.025) were less likely to present with splenomegaly. MICA*A6 carriers were more likely to have thrombocytopenia (p=0.04). Since immunogenetic predisposition is often encoded by complex genetic traits, we have also examined the impacts of HLA class I, II and KIR-ligand/ KIR on clinical presentation of T-LGL. Immunogenetic factors associated with anemia as a predominant feature included HLA-DR7 (p=0.003), Bw4-KIR3DL1 (p=0.03), or having less than 3 inhibitory KIR ligand mismatches (p=0.025). Subjects with HLA-Bw6 (p=0.02) and HLA-A*24 (p=0.05) were more likely to have neutropenia, whereas the patients with HLA*A11 were more likely to have thrombocytopenia (p=0.01). HLA-A*24 (p=0.004), HLA-B*15 (p=0.03), and KIR2DL5 (p=0.045) were associated with poorer clinical response to therapy in contrast to those with HLA*01 (p=0.008) and HLA-B*58 (p=0.045) associated with more favorable clinical responses. In contrast to HLA-B*27 carriers with higher likelihood of splenomegaly (p=0.01), patients with HLA-Bw4-KIR3DL1 (p=0.046), HLA-DR3 (p=0.005), or HLA-DQ2 (p=0.028) were less likely to develop splenomegaly. We did not detect any associations with overall survival. In summary, the overrepresentation of MICA*A5.1 among T-LGL patient had limited impact on clinical outcomes. In contrast to numerous associations between HLA and KIR with clinical presentation of T-LGL, the associations for MICA alleles in our cohort were limited to less likelihood of splenomegaly for MICA*A5.1 and MICA*008 carriers and to higher propensity towards thrombocytopenia for those with MICA*A6. Altogether, our findings extend the evidence for a role of immunogenetic factors in the pathogenesis and clinical outcomes of T-LGL. Disclosures No relevant conflicts of interest to declare.


1987 ◽  
Vol 1 (1) ◽  
pp. 79-81
Author(s):  
Anna Pirelli ◽  
Paola Allavena ◽  
Alessandro Rambaldi ◽  
Maria Di Bello ◽  
Paola Pirovano ◽  
...  

2008 ◽  
Vol 49 (4) ◽  
pp. 828-831 ◽  
Author(s):  
Monika Prochorec-Sobieszek ◽  
Monika Chełstowska ◽  
Grzegorz Rymkiewicz ◽  
Mirosław Majewski ◽  
Krzysztof Warzocha ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1171-1171
Author(s):  
Michael J. Clemente ◽  
Marcin W. Wlodarski ◽  
Aaron D. Viny ◽  
Mohammed Shaik ◽  
Nelli Bejanyan ◽  
...  

Abstract Abstract 1171 T cell large granular lymphocyte leukemia (T-LGLL) is characterized by the chronic proliferation of cytotoxic T lymphocytes (CTL) and is often associated with lineage-restricted cytopenias, autoimmune disease and B cell dyscrasias. In many patients clinical features of a true leukemia are absent. Lack of transformation, absence of progressive increase in blood counts, and a paucity of recurrent chromosomal defects or mutations clearly distinguish LGL from typical leukemic development. Thus, some forms of T-LGLL resemble more of a reactive process and possibly represent an extreme pole of oligo- or polyclonal immune reactions to viruses, autoantigens, or perhaps even exaggerated immune tumor surveillance. We have shown that a highly skewed T cell receptor (TCR) variable beta (Vβ) repertoire is strongly associated with monoclonality of TCR CDR3 regions by sequencing and therefore, detection of a Vβ expansion by flow cytometric clonotyping can serve as a surrogate marker for the presence of a clonal CTL process. Flow cytometric Vβ typing offers an opportunity to study the dynamics of the CTL clonal progression during therapy and throughout the clinical course of disease. We studied 124 patients who not only met the WHO guidelines for the diagnosis of T-LGLL but also show skewing of the Vβ spectrum by flow cytometry consistent with the mono/oligoclonal process persistent for over 6 months; 100% of cases demonstrated both TCRγ rearrangement and abnormal CTL population by flow cytometry. LGL count >900/μL was present in 73% of patients (mean 2513±3571 cells/μL). A pathologic clonal Vβ expansion was defined as > mean + 2SD of controls (n=65). Expansions identified by the Vβ panel were present in 92% (mean clone size 55±28%) and 2% had a borderline expansion (within 20% of 2SD) and 3% a δ/γ CD8+ TCR expansion. In 6% the Vβ expanded clone expressed CD4. Absolute clone count (ACC) was calculated by the Vβ contribution multiplied by the absolute CD8 (or CD4) count per μL of blood. ACC correlated well with LGL count (p<.0001, R2=.58). Clinically, patients presented with anemia (25%), neutropenia (9%), pancytopenia (19%), thrombocytopenia (3%), or multi-lineage cytopenia (29%), and 15% of patients were asymptomatic. In order to assess clonal kinetics, 62 of these patients were available for serial Vβ measurements with a follow up range of 0.5–8 years. Paired statistical analysis of clonal dynamics pre and post therapy (cyclophosphamide, alemtuzumab, methotrexate or cyclosporine) demonstrated a significant decrease in ACC between responders and refractory patients (p=.024). Unexpectedly, some patients displayed a change in the dominant clone as demonstrated by a switch in the major clonal Vβ T cell population, i.e. “clonotype switch.” Overall, 32% exhibited a clonotype switch during the study period, while others exhibited the persistence of multiple clones (22%); in 46% the initially diagnosed Vβ monoclonal expansion persisted. Those with multiple clones were more likely to change clonal dominance (p=.05). Clonotype switch was observed in both in relapsed and in refractory patients, and also was frequently accompanied by a change in clinical hematologic features. Significant absolute lymphocytosis (>4000 lymphocytes/μL) was present in 34/124 (27%) patients while 66 had normal lymphocyte counts and 24 were lymphopenic. Of the patients followed serially who clonotype switched, only 3/20 (15%) had absolute lymphocytosis, suggesting that 2 distinct subtypes of T-LGLL may exist. Patients with high lymphocyte and LGL counts may represent a true leukemic process, are less likely to have associated autoimmune disease or second malignancy, and their dominant clone is stable, suggestive of a single transformed precursor. In contrast, in patients with clonal CTL expansions not associated with absolute lymphocytosis, clonotype switching is difficult to reconcile with a true autonomous leukemic process. In sum, our results suggest that in a significant proportion of patients with T-LGL leukemia, the propensity for clonal CTL dominance is not inherent to an aberrant molecular event within the abnormal CTL clone but may rather be related to extrinsic chronic antigenic drive and immune dysregulation. Furthermore, our results lend credence to the body of evidence suggesting that T-LGL leukemia may not, in many instances, be a true leukemia, and may best be classified as T-LGL lymphoproliferation. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 8 ◽  
pp. 232470962094130
Author(s):  
Sara Naji Rad ◽  
Behnam Rafiee ◽  
Gagan Raju ◽  
Mahdis Solhjoo ◽  
Prachi Anand

Large granular lymphocyte leukemia (LGL) is a clonal, lymphoproliferative disorder with an indolent disease course. T-cell LGL (T-LGL) is the most common type of LGL driven from T-cell lineage (85%). The coexistence of T-LGL with several types of autoimmune disorders, mostly rheumatoid arthritis (RA), has been reported. Felty’s syndrome (FS) is defined by splenomegaly, low neutrophil count, and destructive arthritis and is usually seen in <1% of patients with RA. About 30% to 40% of patients with FS have been reported to have an expansion of large granulated lymphocytes in the circulation. FS and T-LGL are similar in terms of clinical manifestations, response to immunosuppressive therapy, their smoldering course, and immunogenetic findings, proposing FS and T-LGL with RA might be different aspects of a single disease spectrum. In this article, we present a case with long-standing RA who had never been on DMARD (Disease Modifying Anti-Rheumatic Drugs) treatment found to have constitutional symptoms, neutropenia, and splenomegaly, and the patient was diagnosed with T-LGL.


Sign in / Sign up

Export Citation Format

Share Document