Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
2.
Genes Chromosomes Cancer ; 62(3): 139-151, 2023 03.
Article in English | MEDLINE | ID: mdl-36412977

ABSTRACT

Myelodysplastic syndromes (MDS) are hematological malignancies classically defined by the presence of cytopenia(s) and dysmorphic myeloid cells. It is now known that MDS can be preceded by a pre-malignant condition called clonal cytopenia of unknown significance (CCUS), which associates a clonality marker with cytopenia in the absence of criteria of dysplasia. However, to date, it is not clear whether chromosomal abnormalities should be considered in the definition of CCUS or if they carry a prognostic impact in CCUS patients. In this study, we analyzed the clinico-biological features and outcomes of 34 patients who presented with one or more cytopenias, an absence of significant dysplasia, and a presence of a chromosomal abnormality (CA). We named this entity chromosomal abnormality with cytopenia of undetermined significance (CACtUS). We show that these patients are slightly older than MDS patients and that they more frequently presented with normocytic anemia. Most CACtUS patients exhibited only one unbalanced CA. The number and type of mutations were comparable between CACtUS patients and MDS patients. Regardless of the cytogenetic abnormality, the clinicobiological characteristics, overall survival, and risk of progression to high-risk (HR) MDS were similar between CACtUS patients and low-risk MDS patients. Thus, we suggest that CACtUS patients can be considered as HR-CCUS and should receive the follow-up regimen recommended for MDS patients.


Subject(s)
Anemia , Chromosome Disorders , Hematologic Neoplasms , Myelodysplastic Syndromes , Humans , Chromosome Aberrations , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Mutation
5.
Leukemia ; 36(5): 1237-1252, 2022 05.
Article in English | MEDLINE | ID: mdl-35354920

ABSTRACT

Despite recent advances in acute myeloid leukemia (AML) molecular characterization and targeted therapies, a majority of AML cases still lack therapeutically actionable targets. In 127 AML cases with unmet therapeutic needs, as defined by the exclusion of ELN favorable cases and of FLT3-ITD mutations, we identified 51 (40%) cases with alterations in RAS pathway genes (RAS+, mostly NF1, NRAS, KRAS, and PTPN11 genes). In 79 homogeneously treated AML patients from this cohort, RAS+ status were associated with higher white blood cell count, higher LDH, and reduced survival. In AML models of oncogenic addiction to RAS-MEK signaling, the MEK inhibitor trametinib demonstrated antileukemic activity in vitro and in vivo. However, the efficacy of trametinib was heterogeneous in ex vivo cultures of primary RAS+ AML patient specimens. From repurposing drug screens in RAS-activated AML cells, we identified pyrvinium pamoate, an anti-helminthic agent efficiently inhibiting the growth of RAS+ primary AML cells ex vivo, preferentially in trametinib-resistant PTPN11- or KRAS-mutated samples. Metabolic and genetic complementarity between trametinib and pyrvinium pamoate translated into anti-AML synergy in vitro. Moreover, this combination inhibited the propagation of RA+ AML cells in vivo in mice, indicating a potential for future clinical development of this strategy in AML.


Subject(s)
Leukemia, Myeloid, Acute , Synthetic Lethal Mutations , Animals , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation , Oxidative Stress , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , fms-Like Tyrosine Kinase 3/metabolism
6.
Blood Adv ; 6(2): 386-398, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34638130

ABSTRACT

Myelodysplastic syndromes (MDS) represent a heterogeneous group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis leading to peripheral cytopenias and in a substantial proportion of cases to acute myeloid leukemia. The deletion of the long arm of chromosome 11, del(11q), is a rare but recurrent clonal event in MDS. Here, we detail the largest series of 113 cases of MDS and myelodysplastic syndromes/myeloproliferative neoplasms (MDS/MPN) harboring a del(11q) analyzed at clinical, cytological, cytogenetic, and molecular levels. Female predominance, a survival prognosis similar to other MDS, a low monocyte count, and dysmegakaryopoiesis were the specific clinical and cytological features of del(11q) MDS. In most cases, del(11q) was isolated, primary and interstitial encompassing the 11q22-23 region containing ATM, KMT2A, and CBL genes. The common deleted region at 11q23.2 is centered on an intergenic region between CADM1 (also known as Tumor Suppressor in Lung Cancer 1) and NXPE2. CADM1 was expressed in all myeloid cells analyzed in contrast to NXPE2. At the functional level, the deletion of Cadm1 in murine Lineage-Sca1+Kit+ cells modifies the lymphoid-to-myeloid ratio in bone marrow, although not altering their multilineage hematopoietic reconstitution potential after syngenic transplantation. Together with the frequent simultaneous deletions of KMT2A, ATM, and CBL and mutations of ASXL1, SF3B1, and CBL, we show that CADM1 may be important in the physiopathology of the del(11q) MDS, extending its role as tumor-suppressor gene from solid tumors to hematopoietic malignancies.


Subject(s)
Cell Adhesion Molecule-1/metabolism , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Animals , Cell Adhesion Molecule-1/genetics , Chromosome Deletion , Chromosomes, Human, Pair 11 , Female , Genes, Tumor Suppressor , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology
7.
Nat Commun ; 12(1): 5044, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34413298

ABSTRACT

Indirect somatic genetic rescue (SGR) of a germline mutation is thought to be rare in inherited Mendelian disorders. Here, we establish that acquired mutations in the EIF6 gene are a frequent mechanism of SGR in Shwachman-Diamond syndrome (SDS), a leukemia predisposition disorder caused by a germline defect in ribosome assembly. Biallelic mutations in the SBDS or EFL1 genes in SDS impair release of the anti-association factor eIF6 from the 60S ribosomal subunit, a key step in the translational activation of ribosomes. Here, we identify diverse mosaic somatic genetic events (point mutations, interstitial deletion, reciprocal chromosomal translocation) in SDS hematopoietic cells that reduce eIF6 expression or disrupt its interaction with the 60S subunit, thereby conferring a selective advantage over non-modified cells. SDS-related somatic EIF6 missense mutations that reduce eIF6 dosage or eIF6 binding to the 60S subunit suppress the defects in ribosome assembly and protein synthesis across multiple SBDS-deficient species including yeast, Dictyostelium and Drosophila. Our data suggest that SGR is a universal phenomenon that may influence the clinical evolution of diverse Mendelian disorders and support eIF6 suppressor mimics as a therapeutic strategy in SDS.


Subject(s)
Mutation , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosomes/genetics , Ribosomes/pathology , Shwachman-Diamond Syndrome/genetics , Shwachman-Diamond Syndrome/pathology , Adolescent , Adult , Animals , Biological Phenomena , Cells, Cultured , Child , Child, Preschool , Dictyostelium , Drosophila , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Germ Cells , Humans , Infant , Molecular Dynamics Simulation , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Protein Binding , Protein Biosynthesis , Proteins/genetics , Proteins/metabolism , Ribonucleoprotein, U5 Small Nuclear/genetics , Ribonucleoprotein, U5 Small Nuclear/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae , Sequence Homology, Amino Acid , Shwachman-Diamond Syndrome/metabolism , Young Adult
8.
Br J Haematol ; 193(1): 72-82, 2021 04.
Article in English | MEDLINE | ID: mdl-33314017

ABSTRACT

A translocation involving the cyclin-dependent kinase 6 (CDK6) gene [t(CDK6)] is a rare but recurrent abnormality in B-cell neoplasms. To further characterise this aberration, we studied 57 cases; the largest series reported to date. Fluorescence in situ hybridisation analysis confirmed the involvement of CDK6 in all cases, including t(2;7)(p11;q21) immunoglobulin kappa locus (IGK)/CDK6 (n = 51), t(7;14)(q21;q32) CDK6/immunoglobulin heavy locus (IGH) (n = 2) and the previously undescribed t(7;14)(q21;q11) CDK6/T-cell receptor alpha locus (TRA)/T-cell receptor delta locus (TRD) (n = 4). In total, 10 patients were diagnosed with chronic lymphocytic leukaemia, monoclonal B-cell lymphocytosis or small lymphocytic lymphoma, and 47 had small B-cell lymphoma (SmBL) including 36 cases of marginal zone lymphoma (MZL; 34 splenic MZLs, one nodal MZL and one bronchus-associated lymphoid tissue lymphoma). In all, 18 of the 26 cytologically reviewed cases of MZL (69%) had an atypical aspect with prolymphocytic cells. Among the 47 patients with MZL/SmBL, CD5 expression was found in 26 (55%) and the tumour protein p53 (TP53) deletion in 22 (47%). The TP53 gene was mutated in 10/30 (33%); the 7q deletion was detected in only one case, and no Notch receptor 2 (NOTCH2) mutations were found. Immunoglobulin heavy-chain variable-region (IGHV) locus sequencing revealed that none harboured an IGHV1-02*04 gene. Overall survival was 82% at 10 years and not influenced by TP53 aberration. Our present findings suggest that most t(CDK6)+ neoplasms correspond to a particular subgroup of indolent marginal zone B-cell lymphomas with distinctive features.


Subject(s)
CD5 Antigens/metabolism , Cyclin-Dependent Kinase 6/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphoma, B-Cell, Marginal Zone/metabolism , Splenic Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Aged, 80 and over , Bronchial Neoplasms/diagnosis , Bronchial Neoplasms/metabolism , Cell Differentiation , Chromosome Aberrations , Female , Genes, p53/genetics , Humans , Immunoglobulin Heavy Chains/metabolism , In Situ Hybridization, Fluorescence/methods , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, B-Cell, Marginal Zone/diagnosis , Lymphoma, B-Cell, Marginal Zone/drug therapy , Male , Middle Aged , Mutation , Phenotype , Survival Analysis , Tertiary Lymphoid Structures/pathology , Translocation, Genetic/genetics , Trisomy/genetics
10.
Blood ; 134(21): 1821-1831, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31527074

ABSTRACT

B-cell prolymphocytic leukemia (B-PLL) is a rare hematological disorder whose underlying oncogenic mechanisms are poorly understood. Our cytogenetic and molecular assessments of 34 patients with B-PLL revealed several disease-specific features and potential therapeutic targets. The karyotype was complex (≥3 abnormalities) in 73% of the patients and highly complex (≥5 abnormalities) in 45%. The most frequent chromosomal aberrations were translocations involving MYC [t(MYC)] (62%), deletion (del)17p (38%), trisomy (tri)18 (30%), del13q (29%), tri3 (24%), tri12 (24%), and del8p (23%). Twenty-six (76%) of the 34 patients exhibited an MYC aberration, resulting from mutually exclusive translocations or gains. Whole-exome sequencing revealed frequent mutations in TP53, MYD88, BCOR, MYC, SF3B1, SETD2, CHD2, CXCR4, and BCLAF1. The majority of B-PLL used the IGHV3 or IGHV4 subgroups (89%) and displayed significantly mutated IGHV genes (79%). We identified 3 distinct cytogenetic risk groups: low risk (no MYC aberration), intermediate risk (MYC aberration but no del17p), and high risk (MYC aberration and del17p) (P = .0006). In vitro drug response profiling revealed that the combination of a B-cell receptor or BCL2 inhibitor with OTX015 (a bromodomain and extra-terminal motif inhibitor targeting MYC) was associated with significantly lower viability of B-PLL cells harboring a t(MYC). We concluded that cytogenetic analysis is a useful diagnostic and prognostic tool in B-PLL. Targeting MYC may be a useful treatment option in this disease.


Subject(s)
Leukemia, Prolymphocytic, B-Cell/genetics , Proto-Oncogene Proteins c-myc/genetics , Tumor Suppressor Protein p53/genetics , Aged , Aged, 80 and over , Chromosome Aberrations , Cytogenetic Analysis , Female , Humans , Male , Middle Aged , Prognosis
11.
Cytometry B Clin Cytom ; 96(3): 215-222, 2019 05.
Article in English | MEDLINE | ID: mdl-30963682

ABSTRACT

BACKGROUND: Compelling evidence has emerged for the relevance of flow cytometry (FC) in the diagnostic work-up of myelodysplastic syndromes (MDS) but due to technical issues, the erythroid lineage has been under investigated, specifically in the therapeutic context. METHODS: Using the "no red cell lysis" method developed to set up the RED-score, we specifically quantified the fraction of CD117/c-KIT-expressing erythroid precursors in a cohort of 144 MDS patients and studied the correlation with response to erythropoiesis-stimulating agents (ESA) in a sub cohort of 63 low-risk MDS patients. RESULTS: We confirmed the previously reported increase in CD117/c-KIT-expressing erythroid precursors in a subset of MDS patients and demonstrated a strong association between a cut off of CD117/c-KIT-expressing erythroid precursors ≥3% and ESA response (P = 0.001), independent of red blood cell requirement. From our observations, we hypothesized that a decrease in CD117/c-KIT-expressing erythroid precursors could be a mechanism of ESA failure. Moreover, the fraction of CD117/c-KIT-expressing erythroid precursors was correlated with progression-free survival in low-risk MDS patients (P = 0.018). In vitro, we demonstrated in an EPO dependent cell line that CD117/c-KIT expression is necessary for cell survival under EPO stimulation. CONCLUSIONS: The quantification of the CD117/c-KIT-expressing erythroid precursors could be proposed as a new theranostic and prognostic marker in MDS treated by ESA. Future studies will be required to determine whether modulating CD117/c-KIT expression and signaling could be used to improve anemia in MDS. © 2019 International Clinical Cytometry Society.


Subject(s)
Erythroid Precursor Cells/drug effects , Erythropoietin/therapeutic use , Hematinics/therapeutic use , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/drug therapy , Proto-Oncogene Proteins c-kit/genetics , Biomarkers/blood , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Erythropoiesis/drug effects , Erythropoiesis/genetics , Erythropoietin/pharmacology , Female , Gene Expression , Hematinics/pharmacology , Humans , Male , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Primary Cell Culture , Prognosis , Progression-Free Survival , Prospective Studies , Proto-Oncogene Proteins c-kit/metabolism , Risk , Theranostic Nanomedicine/methods
14.
Ann Biol Clin (Paris) ; 75(5): 580-588, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28958970

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired hematopoietic stem cell disorder that must be correctly diagnose because it is a chronic disease with a real impact on the quality of life and the survival of the patients. PNH screening of all patients with anemia or thrombosis is not recommended. We report the case of a 71-year-old male patient referred for chronic anemia. Anemia work-up revealed a misunderstood association of a hemolytic anemia with a negative direct antiglobulin test and iron deficiency. The patient exhibits biological signs of intravascular hemolysis, as well as a recent history of two episodes of deep vein thrombosis. Screening for PNH by flow cytometry shown a PNH clone with a size of approximately 33% of the granulocytes and 11% of the red blood cells. An interstitial deletion of the chromosome 13 was found in the medullar karyotype. PNH through chronic intravascular hemolysis induces an urinary iron loss. This is the only cause of hemolytic anemia inducing iron deficiency.


Subject(s)
Anemia, Hemolytic/complications , Anemia, Iron-Deficiency/complications , Hemoglobinuria, Paroxysmal/diagnosis , Venous Thrombosis/complications , Aged , Anemia, Hemolytic/diagnosis , Anemia, Iron-Deficiency/diagnosis , Diagnosis, Differential , Hemoglobinuria, Paroxysmal/complications , Humans , Male , Venous Thrombosis/diagnosis
15.
Haematologica ; 101(6): 732-40, 2016 06.
Article in English | MEDLINE | ID: mdl-26944475

ABSTRACT

UNLABELLED: Gene expression studies have consistently identified a HOXA-overexpressing cluster of T-cell acute lymphoblastic leukemias, but it is unclear whether these constitute a homogeneous clinical entity, and the biological consequences of HOXA overexpression have not been systematically examined. We characterized the biology and outcome of 55 HOXA-positive cases among 209 patients with adult T-cell acute lymphoblastic leukemia uniformly treated during the Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL)-2003 and -2005 studies. HOXA-positive patients had markedly higher rates of an early thymic precursor-like immunophenotype (40.8% versus 14.5%, P=0.0004), chemoresistance (59.3% versus 40.8%, P=0.026) and positivity for minimal residual disease (48.5% versus 23.5%, P=0.01) than the HOXA-negative group. These differences were due to particularly high frequencies of chemoresistant early thymic precursor-like acute lymphoblastic leukemia in HOXA-positive cases harboring fusion oncoproteins that transactivate HOXA Strikingly, the presence of an early thymic precursor-like immunophenotype was associated with marked outcome differences within the HOXA-positive group (5-year overall survival 31.2% in HOXA-positive early thymic precursor versus 66.7% in HOXA-positive non-early thymic precursor, P=0.03), but not in HOXA-negative cases (5-year overall survival 74.2% in HOXA-negative early thymic precursor versus 57.2% in HOXA-negative non-early thymic precursor, P=0.44). Multivariate analysis further revealed that HOXA positivity independently affected event-free survival (P=0.053) and relapse risk (P=0.039) of chemoresistant T-cell acute lymphoblastic leukemia. These results show that the underlying mechanism of HOXA deregulation dictates the clinico-biological phenotype, and that the negative prognosis of early thymic precursor acute lymphoblastic leukemia is exclusive to HOXA-positive patients, suggesting that early treatment intensification is currently suboptimal for therapeutic rescue of HOXA-positive chemoresistant adult early thymic precursor acute lymphoblastic leukemia. TRIAL REGISTRATION: The GRAALL-2003 and -2005 studies were registered at http://www.clinicaltrials.gov as #NCT00222027 and #NCT00327678, respectively.


Subject(s)
Gene Expression , Homeodomain Proteins/genetics , Phenotype , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Thymus Gland/metabolism , Thymus Gland/pathology , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Immunophenotyping , Male , Middle Aged , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Recurrence , Treatment Outcome , Young Adult
16.
Chromosoma ; 125(4): 789-805, 2016 09.
Article in English | MEDLINE | ID: mdl-26685999

ABSTRACT

Nup98 is an important component of the nuclear pore complex (NPC) and also a rare but recurrent target for chromosomal translocation in leukaemogenesis. Nup98 contains multiple cohesive Gly-Leu-Phe-Gly (GLFG) repeats that are critical notably for the formation of intranuclear GLFG bodies. Previous studies have reported the existence of GLFG bodies in cells overexpressing exogenous Nup98 or in a HeLa subline (HeLa-C) expressing an unusual elevated amount of endogenous Nup98. Here, we have analysed the presence of Nup98-containing bodies in several human cell lines. We found that HEp-2, another HeLa subline, contains GLFG bodies that are distinct from those identified in HeLa-C. Rapid amplification of cDNA ends (RACE) revealed that HEp-2 cells express additional truncated forms of Nup98 fused to a non-coding region of chromosome 11q22.1. Cytogenetic analyses using FISH and array-CGH further revealed chromosomal rearrangements that were distinct from those observed in leukaemic cells. Indeed, HEp-2 cells feature a massive amplification of juxtaposed NUP98 and 11q22.1 loci on a chromosome marker derived from chromosome 3. Unexpectedly, minor co-amplifications of NUP98 and 11q22.1 loci were also observed in other HeLa sublines, but on rearranged chromosomes 11. Altogether, this study reveals that distinct genomic rearrangements affecting NUP98 are associated with the formation of GLFG bodies in specific HeLa sublines.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Nuclear Pore Complex Proteins/genetics , Repetitive Sequences, Amino Acid/genetics , Translocation, Genetic/genetics , Caco-2 Cells , Cell Line, Tumor , Comparative Genomic Hybridization , Gene Amplification/genetics , HeLa Cells , Hep G2 Cells , Humans , In Situ Hybridization, Fluorescence , Leukemia/genetics , Nuclear Pore Complex Proteins/metabolism
17.
Clin Gastroenterol Hepatol ; 12(4): 599-608.e1, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24316103

ABSTRACT

BACKGROUND & AIMS: Little is known about intestinal CD4+ T-cell lymphoma; this rare malignancy is misdiagnosed frequently. We evaluated diagnostic criteria and factors that might affect its development and outcome. METHODS: In a retrospective analysis, we analyzed medical records and intestinal specimens from 10 patients diagnosed with intestinal CD4+ T-cell lymphoma among 115 consecutive patients examined for severe enteropathy with villous atrophy. Samples were analyzed by histology, flow cytometry, and comparative genomic hybridization. RESULTS: Small-intestine epithelial and lamina propria tissues from patients who presented with chronic diarrhea and malnutrition had variable levels of infiltration of CD3+ CD4+ T cells. Flow cytometry showed a high frequency of CD4+ intraepithelial cells, which frequently expressed a specific Vß chain. T-cell receptor ß clonality was confirmed by DNA sequencing. Two patients had HLA and serology results compatible with celiac disease and autoimmune enteropathy, respectively. Two patients were found to have antibodies against human T-cell leukemia virus and 2 patients had signs of a recent infection with the herpes viruses. Comparative genomic hybridization analyses showed heterogeneous chromosomal abnormalities. Symptoms were reduced in patients treated with steroids (n = 5), but not in patients given purine analogues or chemotherapy. Antibodies against CD52 produced clinical and histologic responses in 2 of 2 patients, whereas severe adverse effects developed in 1 patient. At the latest follow-up evaluation, all patients were alive. CONCLUSIONS: There is much heterogeneity in the onset and genetic features of intestinal CD4+ T-cell lymphomas, despite their common presentation as indolent lymphoproliferations of the intestinal mucosa. Patients should be treated with steroids, and possibly antibodies against CD52 (for the most aggressive forms of this disorder).


Subject(s)
CD4-Positive T-Lymphocytes/pathology , Intestinal Neoplasms/etiology , Intestinal Neoplasms/pathology , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/pathology , Aged , Flow Cytometry , Histocytochemistry , Humans , Male , Middle Aged , Nucleic Acid Hybridization , Retrospective Studies , Young Adult
18.
Blood ; 122(23): 3713-22, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24089328

ABSTRACT

Monogenic interleukin-10 (IL-10) and IL-10 receptor (IL-10R) deficiencies cause very early onset severe inflammatory bowel disease. Here, we report that 5 patients with an IL-10R1 (n = 1) or IL-10R2 (n = 4) deficiency developed B-cell non-Hodgkin lymphoma between the ages of 5 and 6 years (which was recurrent in 1 patient). These lymphomas had some of the characteristics of diffuse large B-cell lymphomas and contained monoclonal, Epstein-Barr virus-negative germinal center B cells. The tumors displayed a remarkably homogeneous signature, with original activation of the nuclear factor κB pathway and a decrease in intratumor T-cell infiltration. Hence, IL-10R deficiency is associated with a high risk of developing B-cell lymphoma. Our results revealed an unexpected role of the IL-10R pathway in lymphomagenesis.


Subject(s)
Interleukin-10 Receptor alpha Subunit/deficiency , Interleukin-10 Receptor alpha Subunit/genetics , Interleukin-10 Receptor beta Subunit/deficiency , Interleukin-10 Receptor beta Subunit/genetics , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Adolescent , Age of Onset , Child , Child, Preschool , Female , Gene Expression Profiling , Genes, rel , Genetic Predisposition to Disease , Germinal Center/immunology , Germinal Center/pathology , Humans , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Interleukin-10/metabolism , Lymphoma, B-Cell/etiology , Lymphoma, Large B-Cell, Diffuse/etiology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Male , Mutation , NF-kappa B/metabolism , Pedigree , Signal Transduction
19.
Blood ; 122(20): 3521-32, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24014241

ABSTRACT

Cancer cells require nutrients and energy to adapt to increased biosynthetic activity, and protein synthesis inhibition downstream of mammalian target of rapamycin complex 1 (mTORC1) has shown promise as a possible therapy for acute myeloid leukemia (AML). Glutamine contributes to leucine import into cells, which controls the amino acid/Rag/mTORC1 signaling pathway. We show in our current study that glutamine removal inhibits mTORC1 and induces apoptosis in AML cells. The knockdown of the SLC1A5 high-affinity transporter for glutamine induces apoptosis and inhibits tumor formation in a mouse AML xenotransplantation model. l-asparaginase (l-ase) is an anticancer agent also harboring glutaminase activity. We show that l-ases from both Escherichia coli and Erwinia chrysanthemi profoundly inhibit mTORC1 and protein synthesis and that this inhibition correlates with their glutaminase activity levels and produces a strong apoptotic response in primary AML cells. We further show that l-ases upregulate glutamine synthase (GS) expression in leukemic cells and that a GS knockdown enhances l-ase-induced apoptosis in some AML cells. Finally, we observe a strong autophagic process upon l-ase treatment. These results suggest that l-ase anticancer activity and glutamine uptake inhibition are promising new therapeutic strategies for AML.


Subject(s)
Glutamine/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Adult , Aged , Aged, 80 and over , Amino Acid Transport System ASC/antagonists & inhibitors , Amino Acid Transport System ASC/genetics , Animals , Apoptosis/drug effects , Asparaginase/isolation & purification , Asparaginase/pharmacology , Autophagy/drug effects , Bacterial Proteins/pharmacology , Biological Transport/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Dickeya chrysanthemi/enzymology , Drug Screening Assays, Antitumor , Escherichia coli Proteins/pharmacology , Female , Glutaminase/isolation & purification , Glutaminase/pharmacology , Glutamine/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Acute/drug therapy , Leukemia, Myelomonocytic, Acute/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Nude , Middle Aged , Minor Histocompatibility Antigens , Multiprotein Complexes/antagonists & inhibitors , Protein Biosynthesis/drug effects , RNA Interference , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays , Young Adult
20.
Cancer Genet ; 206(5): 162-73, 2013 May.
Article in English | MEDLINE | ID: mdl-23827691

ABSTRACT

Chromosomal translocations involving the immunoglobulin heavy chain locus (IGH@) are recurrent but rare in B-cell precursor acute lymphoblastic leukemia (BCP-ALL), and various partner genes have been described. Here, we report a new series of 29 cases of BCP-ALL with IGH@ translocations. The partner gene was identified by fluorescence in situ hybridization and/or molecular cloning in 20 patients. Members of the CEBP gene family (n = 11), BCL2 (n = 3), ID4 (n = 3), EPOR (n = 2), and TRA/D@ (n = 1) were identified and demonstrated by quantitative real-time reverse transcriptase-polymerase chain reaction to be markedly up-regulated. The present cases, added to those already reported, confirm the diversity of the partner genes, which, apart from BCL2, are specific to BCP-ALL. Collectively, patients with IGH@ translocations may represent a novel sub-group of BCP-ALL occurring in adolescents and young adults.


Subject(s)
Immunoglobulin Heavy Chains/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Translocation, Genetic , Adolescent , Adult , Aged , Aged, 80 and over , CCAAT-Enhancer-Binding Proteins/genetics , Child , Child, Preschool , Chromosomes, Human, Pair 14 , Cloning, Molecular , Female , Humans , In Situ Hybridization, Fluorescence , Inhibitor of Differentiation Proteins/genetics , Male , Middle Aged , Proto-Oncogene Proteins c-bcl-2/genetics , Real-Time Polymerase Chain Reaction , Receptors, Erythropoietin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...