Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Invest New Drugs ; 38(5): 1598-1600, 2020 10.
Article in English | MEDLINE | ID: mdl-31965420

ABSTRACT

B cell prolymphocytic leukemia (B-PLL) is a rare and aggressive disease that is associated with poor survival. Although initially asymptomatic patients do not require therapy, most patients will progress and inevitably require treatment. More than 50% of patients with B-PLL carry abnormalities in the TP53 tumor suppressor gene and/or complex karyotype and show resistance to conventional chemotherapy. The efficacy of ibrutinib, a B cell receptor inhibitor, for B-PLL with the TP53 abnormality as second-line therapy was recently demonstrated. We herein report that low-dose ibrutinib as upfront therapy induced a complete response in a B-PLL patient with the TP53 abnormality, whose condition has since remained stable with no recurrence for 12 months. Effective treatments for B-PLL are lacking and given its rarity, prospective comparative therapies are not yet available. This case suggests that upfront therapy with ibrutinib improves the outcome of B-PLL.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/therapeutic use , Leukemia, Prolymphocytic, B-Cell/drug therapy , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Adenine/therapeutic use , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Aged , Humans , Leukemia, Prolymphocytic, B-Cell/genetics , Male , Mutation , Treatment Outcome , Tumor Suppressor Protein p53/genetics
2.
Best Pract Res Clin Haematol ; 32(3): 217-228, 2019 09.
Article in English | MEDLINE | ID: mdl-31585622

ABSTRACT

Prolymphocytic leukaemias B-PLL and T-PLL are rare disorders, typically with an aggressive clinical course and poor prognosis. Combining morphology, immunophenotyping, cytogenetic and molecular diagnostics reliably separates B-PLL and T-PLL from one another and other disorders. In T-PLL discovery of frequent mutations in the JAK-STAT pathway have increased understanding of disease pathogenesis. Alemtuzumab (anti-CD52) produces excellent response rates but long-term remissions are only achieved in a minority following consolidation with allogeneic stem cell transplant. Molecular abnormalities in B-PLL are less understood. Disruption of TP53 is a key finding, conveying chemotherapy resistance requiring novel therapies such as B-cell receptor inhibitors (BCRi). Both conditions require improved pathobiological knowledge to identify new treatment targets and guide therapy with novel pathway inhibitors.


Subject(s)
Alemtuzumab/therapeutic use , Hematopoietic Stem Cell Transplantation , Leukemia, Prolymphocytic, B-Cell , Leukemia, Prolymphocytic, T-Cell , Humans , Leukemia, Prolymphocytic, B-Cell/genetics , Leukemia, Prolymphocytic, B-Cell/metabolism , Leukemia, Prolymphocytic, B-Cell/pathology , Leukemia, Prolymphocytic, B-Cell/therapy , Leukemia, Prolymphocytic, T-Cell/genetics , Leukemia, Prolymphocytic, T-Cell/metabolism , Leukemia, Prolymphocytic, T-Cell/pathology , Leukemia, Prolymphocytic, T-Cell/therapy
3.
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
6.
Curr Oncol Rep ; 19(4): 29, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28324286

ABSTRACT

PURPOSE OF REVIEW: We aimed to produce a comprehensive update on clinical and biological data regarding two rare lymphoid neoplasms, B and T prolymphocytic leukemias, and assess therapeutic management in the light of new molecular insights and the advent of targeted therapies. RECENT FINDINGS: B cell prolymphocytic leukemia (B-PLL) diagnosis remains challenging in the absence of clear immunophenotypic or cytogenetic signature and overlap with mantle cell lymphoma. New molecular defects have been identified in T cell prolymphocytic leukemia (T-PLL), especially in the JAK STAT pathway. Like in chronic lymphocytic leukemia (CLL), B-PLL treatment depends on the presence of TP53 dysfunction. In T-PLL, alemtuzumab still remains the standard of care. Allogeneic transplantation is the only curable option. Thanks to reduced intensity conditioning regimens, it has become accessible to a larger number of patients. PLL prognosis remains poor with conventional therapies. However, great advances in the understanding of both T- and B-PLL pathogenesis lead to promising new therapeutic agents.


Subject(s)
Leukemia, Prolymphocytic, B-Cell/diagnosis , Leukemia, Prolymphocytic, B-Cell/genetics , Leukemia, Prolymphocytic, B-Cell/therapy , Leukemia, Prolymphocytic, T-Cell/diagnosis , Alemtuzumab , Antibodies, Monoclonal, Humanized/therapeutic use , Humans , Immunophenotyping/methods , Leukemia, Prolymphocytic, T-Cell/genetics , Leukemia, Prolymphocytic, T-Cell/therapy , Transplantation, Homologous/methods
8.
Epigenetics ; 11(6): 449-55, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27128508

ABSTRACT

Based on the methylation status of 5 single CpG sites, a novel epigenetic classification of chronic lymphocytic leukemia (CLL) was recently proposed, classifying CLL patients into 3 clinico-biological subgroups with different outcome, termed memory like CLL (m-CLL), naïve like CLL (n-CLL), and a third intermediate CLL subgroup (i-CLL). While m-CLL and n-CLL patients at large corresponded to patients carrying mutated and unmutated IGHV genes, respectively, limited information exists regarding the less defined i-CLL group. Using pyrosequencing, we investigated the prognostic impact of the proposed 5 CpG signature in a well-characterized CLL cohort (135 cases), including IGHV-mutated and unmutated patients as well as clinically aggressive stereotyped subset #2 patients. Overall, we confirmed the signature's association with established prognostic markers. Moreover, in the presence of the IGHV mutational status, the epigenetic signature remained independently associated with both time-to-first-treatment and overall survival in multivariate analyses. As a prime finding, we observed that subset #2 patients were predominantly classified as i-CLL, probably reflecting their borderline IGHV mutational status (97-99% germline identity), though having a similarly poor prognosis as n-CLL patients. In summary, we validated the epigenetic classifier as an independent factor in CLL prognostication and provide further evidence that subset #2 is a member of the i-CLL group, hence supporting the existence of a third, intermediate epigenetic subgroup.


Subject(s)
Biomarkers, Tumor/standards , DNA Methylation , Epigenesis, Genetic , Leukemia, Prolymphocytic, B-Cell/classification , Adult , Aged , Biomarkers, Tumor/genetics , Female , Humans , Leukemia, Prolymphocytic, B-Cell/diagnosis , Leukemia, Prolymphocytic, B-Cell/genetics , Male , Middle Aged , Mutation , Prognosis , Survival Analysis
9.
J Clin Exp Hematop ; 54(3): 219-24, 2014.
Article in English | MEDLINE | ID: mdl-25501113

ABSTRACT

An 80-year-old man was referred to our department because of lymphocytosis. His white cell count was 17.1 × 10(3)/µL, with 64% prolymphocytes. He did not exhibit splenomegaly or lymphadenopathy. Prolymphocytes were CD5(+), CD10(-), CD19(+), CD20(+), CD21(+weak), CD22(+), CD23(-), and HLA-DR(+), and expressed µÎ´/λ cell-surface immunoglobulins. G-banding and fluorescence in situ hybridization using c-MYC and immunoglobulin heavy-chain (IgH) gene probe revealed that leukemia cells carried the t(8;14)(q24;q32)/c-MYC-IgH fusion gene, and breakage and reunion occurred within the non-coding region of c-MYC exon 1 as well as the α switch region of IgH. Nine months after the initial presentation, the patient's hemoglobin level fell to 5.7 g/dL. Coombs' test was positive and marked hypoplasia of erythroid precursors was detected in his bone marrow. The patient was treated with prednisolone followed by 4 weekly doses of rituximab, which led to resolution of the anemia and complete response of the underlying leukemia. The role of t(8;14)(q24;q32)/c-MYC-IgH in the pathogenesis of B-cell prolymphocytic leukemia (B-PLL) may not be identical to that in aggressive lymphoid neoplasms, and, in the present case, autoantibodies targeting both mature red cells and erythroid precursors may have been concurrently produced in the setting of B-PLL.


Subject(s)
Anemia, Hemolytic, Autoimmune/genetics , Leukemia, Prolymphocytic, B-Cell/blood , Leukemia, Prolymphocytic, B-Cell/genetics , Red-Cell Aplasia, Pure/genetics , Aged, 80 and over , Anemia, Hemolytic, Autoimmune/blood , Anemia, Hemolytic, Autoimmune/pathology , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 8 , Humans , Male , Red-Cell Aplasia, Pure/blood , Red-Cell Aplasia, Pure/pathology , Translocation, Genetic
10.
Am J Clin Pathol ; 142(3): 347-54, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25125625

ABSTRACT

OBJECTIVES: B-cell prolymphocytic leukemia (B-PLL) remains a controversial entity, and its molecular pathogenesis is largely unknown. Patients are older, typically having marked lymphocytosis and splenomegaly in the absence of lymphadenopathy. It is defined as a mature B-cell leukemia with more than 55% circulating prolymphocytes. Leukemic mantle cell lymphoma and chronic lymphocytic leukemia in prolymphocytic transformation must be excluded. METHODS: Case archives were retrospectively reviewed for B-PLL in patients without a previous diagnosis of chronic lymphocytic leukemia or other B-cell neoplasm. RESULTS: We identified six cases of B-PLL with available cytogenetic data, five of which showed evidence of aberrations in MYC. Three cases showed additional signals for the MYC gene by fluorescence in situ hybridization (FISH), and two cases demonstrated t(8;14)MYC/IGH by karyotyping or FISH. High levels of MYC protein expression were detected in all cases tested with MYC aberrations. CONCLUSIONS: These results suggest that deregulation of MYC plays an important role in the pathogenesis of B-PLL and expands the spectrum of B-cell neoplasms associated with aberrations of MYC.


Subject(s)
Leukemia, Prolymphocytic, B-Cell/genetics , Leukocytosis/genetics , Proto-Oncogene Proteins c-myc/genetics , Aged , Aged, 80 and over , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Leukemia, Prolymphocytic, B-Cell/pathology , Leukocytosis/pathology , Lymphocytes/pathology , Male , Middle Aged , Retrospective Studies
11.
Blood ; 124(3): 412-9, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24891323

ABSTRACT

B-cell prolymphocytic leukemia (B-PLL) is a rare mature B-cell malignancy that may be hard to distinguish from mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL). B-PLL cases with a t(11;14) were redefined as MCL in the World Health Organization 2008 classification. We evaluated 13 B-PLL patients [7 being t(11;14)-positive (B-PLL+) and 6 negative (B-PLL-)] and compared them with MCL and CLL patients. EuroFlow-based immunophenotyping showed significant overlap between B-PLL+ and B-PLL-, as well as between B-PLL and MCL, whereas CLL clustered separately. Immunogenotyping showed specific IGHV gene usage partly resembling MCL. Gene expression profiling showed no separation between B-PLL+ and B-PLL- but identified 3 subgroups. One B-PLL subgroup clustered close to CLL and another subgroup clustered with leukemic MCL; both were associated with prolonged survival. A third subgroup clustered close to nodal MCL and was associated with short survival. Gene expression profiles of both B-PLL+ and B-PLL- showed best resemblance with normal immunoglobulin M-only B-cells. Our data confirm that B-PLL+ is highly comparable to MCL, indicate that B-PLL- also may be considered as a specific subgroup of MCL, and suggest that B-PLL is part of a spectrum, ranging from CLL-like B-PLL, to leukemic MCL-like B-PLL, to nodal MCL-like B-PLL.


Subject(s)
Leukemia, Prolymphocytic, B-Cell/classification , Lymphoma, Mantle-Cell/classification , Adult , Aged , B-Lymphocyte Subsets/immunology , Female , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, Immunoglobulin Heavy Chain , Humans , Immunophenotyping , Leukemia, Lymphocytic, Chronic, B-Cell/classification , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Prolymphocytic, B-Cell/genetics , Leukemia, Prolymphocytic, B-Cell/immunology , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/immunology , Male , Middle Aged , Transcriptome
12.
Hum Pathol ; 43(11): 1828-38, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22520947

ABSTRACT

We describe 4 patients aged 62 to 79 years with splenomegaly and bone marrow involvement by splenic B-cell lymphoma who developed more than 55% prolymphocytes in blood. The diagnosis of B-cell prolymphocytic leukemia was considered clinically based on a markedly elevated leukocyte (up to 131.5×10(9)/L) or prolymphocyte (up to 86%) count. Splenectomy was performed in all patients, and spleen weight ranged from 1500 to 2380 g. In 3 patients, the neoplasms were classified as splenic marginal zone lymphoma, and in 1 patient, the neoplasm was classified as splenic diffuse red pulp small B-cell lymphoma. In 2 patients, splenectomy preceded a B-cell prolymphocytic leukemia-like picture, and the spleen showed splenic marginal zone lymphoma or splenic diffuse red pulp small B-cell lymphoma with increased (10%-20%) nucleolated cells consistent with prolymphocytes. In the other 2 patients, a B-cell prolymphocytic leukemia-like picture prompted splenectomy. Initial examination of bone marrow in these patients suggested splenic marginal zone lymphoma. The spleen specimens showed extensive involvement by splenic marginal zone lymphoma with numerous prolymphocytes. Flow cytometry immunophenotyping in all cases showed lymphocytes positive for monotypic surface immunoglobulin (bright), pan-B-cell antigens, CD11c, CD22, and FMC7. Immunohistochemical analysis in all patients showed moderate to bright p53 expression in the spleen (n=3) or bone marrow (n=2). Annexin A1 and cyclin D1 were negative in all cases. Conventional cytogenetic analysis showed del(7q) in 3 patients. We conclude that splenic B-cell lymphoma of various types can undergo prolymphocytoid transformation with more than 55% prolymphocytes in the blood mimicking B-cell prolymphocytic leukemia.


Subject(s)
Cell Transformation, Neoplastic , Leukemia, Prolymphocytic, B-Cell/pathology , Lymphoma, B-Cell, Marginal Zone/pathology , Splenic Neoplasms/pathology , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Marrow Cells/pathology , Chromosome Deletion , Chromosomes, Human, Pair 7 , Combined Modality Therapy , Diagnosis, Differential , Female , Flow Cytometry , Humans , Immunophenotyping , In Situ Hybridization, Fluorescence , Leukemia, Prolymphocytic, B-Cell/genetics , Leukemia, Prolymphocytic, B-Cell/metabolism , Leukemia, Prolymphocytic, B-Cell/therapy , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, B-Cell, Marginal Zone/metabolism , Lymphoma, B-Cell, Marginal Zone/therapy , Male , Middle Aged , Splenectomy , Splenic Neoplasms/genetics , Splenic Neoplasms/metabolism , Splenic Neoplasms/therapy , Tumor Suppressor Protein p53/metabolism
13.
Invest New Drugs ; 30(1): 417-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20953816

ABSTRACT

The multi-kinase inhibitor dasatinib induced a variable but significant decrease of viability in both p53(wild-type) (EHEB, JVM-2, JVM-3) and p53(mutated) (MEC-1, MEC-2, BJAB) prolymphocytic B leukemic cells, due to a combination of cell cycle block in G1 and apoptosis. Antibody phospho-kinase array analysis revealed that dasatinib inhibited the phosphorylation of various kinases, including ERK1/2 and p38/MAPK as well as of STAT3 transcription factors, in both p53(wild-type) and p53(mutated) cells. Therefore, dasatinib might offer a novel therapeutic strategy not only for p53(wild-type), but also for p53(mutated) B malignancies that have the worst prognosis and urgently need innovative therapeutic approaches.


Subject(s)
Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , Granulocyte Precursor Cells/drug effects , Leukemia, Prolymphocytic, B-Cell/metabolism , Mutation , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dasatinib , G1 Phase Cell Cycle Checkpoints/drug effects , Granulocyte Precursor Cells/metabolism , Granulocyte Precursor Cells/pathology , Humans , Leukemia, Prolymphocytic, B-Cell/genetics , Leukemia, Prolymphocytic, B-Cell/pathology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Time Factors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Blood ; 118(8): 2184-90, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21730354

ABSTRACT

Forodesine and nelarabine (the pro-drug of ara-G) are 2 nucleoside analogues with promising anti-leukemic activity. To better understand which pediatric patients might benefit from forodesine or nelarabine (ara-G) therapy, we investigated the in vitro sensitivity to these drugs in 96 diagnostic pediatric leukemia patient samples and the mRNA expression levels of different enzymes involved in nucleoside metabolism. Forodesine and ara-G cytotoxicities were higher in T-cell acute lymphoblastic leukemia (T-ALL) samples than in B-cell precursor (BCP)-ALL and acute myeloid leukemia (AML) samples. Resistance to forodesine did not preclude ara-G sensitivity and vice versa, indicating that both drugs rely on different resistance mechanisms. Differences in sensitivity could be partly explained by significantly higher accumulation of intracellular dGTP in forodesine-sensitive samples compared with resistant samples, and higher mRNA levels of dGK but not dCK. The mRNA levels of the transporters ENT1 and ENT2 were higher in ara-G-sensitive than -resistant samples. We conclude that especially T-ALL, but also BCP-ALL, pediatric patients may benefit from forodesine or nelarabine (ara-G) treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Prolymphocytic, B-Cell/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Prodrugs/therapeutic use , Purine Nucleosides/therapeutic use , Pyrimidinones/therapeutic use , Cell Line, Tumor , Child , Deoxycytidine Kinase/genetics , Deoxyguanine Nucleotides/metabolism , Drug Resistance, Neoplasm , Equilibrative Nucleoside Transporter 1/genetics , Equilibrative-Nucleoside Transporter 2/genetics , Gene Expression , Humans , In Vitro Techniques , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Prolymphocytic, B-Cell/genetics , Leukemia, Prolymphocytic, B-Cell/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Purines/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
16.
J Exp Med ; 206(8): 1739-53, 2009 Aug 03.
Article in English | MEDLINE | ID: mdl-19620627

ABSTRACT

B cell lineage acute lymphoblastic leukemia (ALL) arises in virtually all cases from B cell precursors that are arrested at pre-B cell receptor-dependent stages. The Philadelphia chromosome-positive (Ph(+)) subtype of ALL accounts for 25-30% of cases of adult ALL, has the most unfavorable clinical outcome among all ALL subtypes and is defined by the oncogenic BCR-ABL1 kinase and deletions of the IKAROS gene in >80% of cases. Here, we demonstrate that the pre-B cell receptor functions as a tumor suppressor upstream of IKAROS through induction of cell cycle arrest in Ph(+) ALL cells. Pre-B cell receptor-mediated cell cycle arrest in Ph(+) ALL cells critically depends on IKAROS function, and is reversed by coexpression of the dominant-negative IKAROS splice variant IK6. IKAROS also promotes tumor suppression through cooperation with downstream molecules of the pre-B cell receptor signaling pathway, even if expression of the pre-B cell receptor itself is compromised. In this case, IKAROS redirects oncogenic BCR-ABL1 tyrosine kinase signaling from SRC kinase-activation to SLP65, which functions as a critical tumor suppressor downstream of the pre-B cell receptor. These findings provide a rationale for the surprisingly high frequency of IKAROS deletions in Ph(+) ALL and identify IKAROS-mediated cell cycle exit as the endpoint of an emerging pathway of pre-B cell receptor-mediated tumor suppression.


Subject(s)
Ikaros Transcription Factor/physiology , Leukemia, Prolymphocytic, B-Cell/genetics , Philadelphia Chromosome , Pre-B Cell Receptors/physiology , Adaptor Proteins, Signal Transducing/physiology , Adult , Animals , Cell Cycle , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Down-Regulation , Gene Deletion , Genes, abl , Humans , Ikaros Transcription Factor/deficiency , Ikaros Transcription Factor/genetics , Leukemia, Prolymphocytic, B-Cell/pathology , Leukemia, Prolymphocytic, B-Cell/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Pre-B Cell Receptors/deficiency , Pre-B Cell Receptors/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL