Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Am Chem Soc ; 146(19): 13317-13325, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38700457

ABSTRACT

We describe the synthesis and biological testing of ruthenium-bipyridine ruxolitinib (RuBiRuxo), a photoreleasable form of ruxolitinib, a JAK inhibitor used as an antitumoral agent in cutaneous T-cell lymphomas (CTCL). This novel caged compound is synthesized efficiently, is stable in aqueous solution at room temperature, and is photoreleased rapidly by visible light. Irradiation of RuBiRuxo reduces cell proliferation and induces apoptosis in a light- and time-dependent manner in a CTCL cell line. This effect is specific and is mediated by a decreased phosphorylation of STAT proteins. Our results demonstrate the potential of ruthenium-based photocompounds and light-based therapeutic approaches for the potential treatment of cutaneous lymphomas and other pathologies.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Proliferation , Nitriles , Pyrazoles , Pyrimidines , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Nitriles/chemistry , Nitriles/pharmacology , Nitriles/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/chemical synthesis , Apoptosis/drug effects , Pyrazoles/pharmacology , Pyrazoles/chemistry , Pyrazoles/chemical synthesis , Cell Line, Tumor , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/chemical synthesis , Ruthenium/chemistry , Ruthenium/pharmacology , Light , Molecular Structure , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism
2.
J Invest Dermatol ; 2024 Jan 14.
Article in English | MEDLINE | ID: mdl-38219917

ABSTRACT

Cutaneous T-cell lymphomas are mature lymphoid neoplasias resulting from the malignant transformation of skin-resident T-cells. A distinctive clinical feature of cutaneous T-cell lymphomas is their sensitivity to treatment with histone deacetylase inhibitors. However, responses to histone deacetylase inhibitor therapy are universally transient and noncurative, highlighting the need for effective and durable drug combinations. In this study, we demonstrate that the combination of romidepsin, a selective class I histone deacetylase inhibitor, with afatinib, an EGFR family inhibitor, induces strongly synergistic antitumor effects in cutaneous T-cell lymphoma models in vitro and in vivo through abrogation of Jak-signal transducer and activator of transcription signaling. These results support a previously unrecognized potential role for histone deacetylase inhibitor plus afatinib combination in the treatment of cutaneous T-cell lymphomas.

3.
Cancer Discov ; 12(11): 2646-2665, 2022 11 02.
Article in English | MEDLINE | ID: mdl-35984649

ABSTRACT

Low-intensity maintenance therapy with 6-mercaptopurine (6-MP) limits the occurrence of acute lymphoblastic leukemia (ALL) relapse and is central to the success of multiagent chemotherapy protocols. Activating mutations in the 5'-nucleotidase cytosolic II (NT5C2) gene drive resistance to 6-MP in over 35% of early relapse ALL cases. Here we identify CRCD2 as a first-in-class small-molecule NT5C2 nucleotidase inhibitor broadly active against leukemias bearing highly prevalent relapse-associated mutant forms of NT5C2 in vitro and in vivo. Importantly, CRCD2 treatment also enhanced the cytotoxic activity of 6-MP in NT5C2 wild-type leukemias, leading to the identification of NT5C2 Ser502 phosphorylation as a novel NT5C2-mediated mechanism of 6-MP resistance in this disease. These results uncover an unanticipated role of nongenetic NT5C2 activation as a driver of 6-MP resistance in ALL and demonstrate the potential of NT5C2 inhibitor therapy for enhancing the efficacy of thiopurine maintenance therapy and overcoming resistance at relapse. SIGNIFICANCE: Relapse-associated NT5C2 mutations directly contribute to relapse in ALL by driving resistance to chemotherapy with 6-MP. Pharmacologic inhibition of NT5C2 with CRCD2, a first-in-class nucleotidase inhibitor, enhances the cytotoxic effects of 6-MP and effectively reverses thiopurine resistance mediated by genetic and nongenetic mechanisms of NT5C2 activation in ALL. This article is highlighted in the In This Issue feature, p. 2483.


Subject(s)
Antineoplastic Agents , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Mercaptopurine/pharmacology , Mercaptopurine/therapeutic use , 5'-Nucleotidase/genetics , 5'-Nucleotidase/therapeutic use , Drug Resistance, Neoplasm/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antineoplastic Agents/therapeutic use , Recurrence
4.
Cell Rep ; 39(3): 110695, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35443168

ABSTRACT

Peripheral T cell lymphoma not otherwise specified (PTCL-NOS) comprises heterogeneous lymphoid malignancies characterized by pleomorphic lymphocytes and variable inflammatory cell-rich tumor microenvironment. Genetic drivers in PTCL-NOS include genomic alterations affecting the VAV1 oncogene; however, their specific role and mechanisms in PTCL-NOS remain incompletely understood. Here we show that expression of Vav1-Myo1f, a recurrent PTCL-associated VAV1 fusion, induces oncogenic transformation of CD4+ T cells. Notably, mouse Vav1-Myo1f lymphomas show T helper type 2 features analogous to high-risk GATA3+ human PTCL. Single-cell transcriptome analysis reveals that Vav1-Myo1f alters T cell differentiation and leads to accumulation of tumor-associated macrophages (TAMs) in the tumor microenvironment, a feature linked with aggressiveness in human PTCL. Importantly, therapeutic targeting of TAMs induces strong anti-lymphoma effects, highlighting the lymphoma cells' dependency on the microenvironment. These results demonstrate an oncogenic role for Vav1-Myo1f in the pathogenesis of PTCL, involving deregulation in T cell polarization, and identify the lymphoma-associated macrophage-tumor microenvironment as a therapeutic target in PTCL.


Subject(s)
Lymphoma, T-Cell, Peripheral , Animals , Gene Fusion , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/metabolism , Lymphoma, T-Cell, Peripheral/pathology , Macrophages/metabolism , Mice , Myosin Type I/genetics , Oncogenes , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/metabolism , Tumor Microenvironment/genetics
5.
Nat Aging ; 2(11): 1008-1023, 2022 11.
Article in English | MEDLINE | ID: mdl-37118089

ABSTRACT

Aging is characterized by an accumulation of myeloid-biased hematopoietic stem cells (HSCs) with reduced developmental potential. Genotoxic stress and epigenetic alterations have been proposed to mediate age-related HSC loss of regenerative and self-renewal potential. However, the mechanisms underlying these changes remain largely unknown. Genetic inactivation of the plant homeodomain 6 (Phf6) gene, a nucleolar and chromatin-associated factor, antagonizes age-associated HSC decline. Immunophenotyping, single-cell transcriptomic analyses and transplantation assays demonstrated markedly decreased accumulation of immunophenotypically defined HSCs, reduced myeloid bias and increased hematopoietic reconstitution capacity with preservation of lymphoid differentiation potential in Phf6-knockout HSCs from old mice. Moreover, deletion of Phf6 in aged mice rejuvenated immunophenotypic, transcriptional and functional hallmarks of aged HSCs. Long-term HSCs from old Phf6-knockout mice showed epigenetic rewiring and transcriptional programs consistent with decreased genotoxic stress-induced HSC aging. These results identify Phf6 as an important epigenetic regulator of HSC aging.


Subject(s)
Aging , Hematopoietic Stem Cells , Mice , Animals , Mice, Knockout , Aging/genetics , Cell Differentiation , Epigenesis, Genetic , Repressor Proteins/genetics
6.
J Invest Dermatol ; 141(12): 2908-2920.e7, 2021 12.
Article in English | MEDLINE | ID: mdl-34089720

ABSTRACT

Sézary syndrome is an aggressive and disseminated form of cutaneous T-cell lymphoma associated with dismal prognosis in which the histone deacetylase inhibitor romidepsin has shown remarkable activity as a single agent. However, clinical responses to romidepsin are typically transient, highlighting the need for more effective therapies. In this study, we show synergistic antilymphoma effects of romidepsin in combination with mechlorethamine, an alkylating agent, in cutaneous T-cell lymphoma cell lines and primary samples with strong antitumor effects in an in vivo model of Sézary syndrome. Mechanistically, gene expression profiling points to abrogation of Jak/signal transducer and activator of transcription (STAT) signaling as an important mediator of this interaction. Consistently, the combination of mechlorethamine plus romidepsin resulted in downregulation of STAT5 phosphorylation in romidepsin-sensitive cell lines and primary Sézary syndrome samples, but not in romidepsin-resistant tumors. Moreover, in further support of Jak/STAT signaling as a modulator of romidepsin activity in cutaneous T-cell lymphoma, treatment with romidepsin in combination with Jak inhibitors resulted in markedly increased therapeutic responses. Overall, these results support a role for romidepsin plus mechlorethamine in combination in the treatment of cutaneous T-cell lymphoma and uncover a previously unrecognized role for Jak/STAT signaling in the response to romidepsin and romidepsin-based combination therapies in Sézary syndrome.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Depsipeptides/administration & dosage , Janus Kinase Inhibitors/pharmacology , Lymphoma, T-Cell, Cutaneous/drug therapy , Mechlorethamine/administration & dosage , STAT Transcription Factors/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Cell Line, Tumor , Drug Synergism , Humans , Mice , STAT Transcription Factors/physiology , Signal Transduction/drug effects
7.
Nat Cancer ; 2(1): 98-113, 2021 01.
Article in English | MEDLINE | ID: mdl-33928261

ABSTRACT

Angioimmunoblastic T cell lymphoma (AITL) and peripheral T cell lymphoma not-otherwise-specified (PTCL, NOS) have poor prognosis and lack driver actionable targets for directed therapies in most cases. Here we identify FYN-TRAF3IP2 as a recurrent oncogenic gene fusion in AITL and PTCL, NOS tumors. Mechanistically, we show that FYN-TRAF3IP2 leads to aberrant NF-κB signaling downstream of T cell receptor activation. Consistent with a driver oncogenic role, FYN-TRAF3IP2 expression in hematopoietic progenitors induces NF-κB-driven T cell transformation in mice and cooperates with loss of the Tet2 tumor suppressor in PTCL development. Moreover, abrogation of NF-κB signaling in FYN-TRAF3IP2-induced tumors with IκB kinase inhibitors delivers strong anti-lymphoma effects in vitro and in vivo. These results demonstrate an oncogenic and pharmacologically targetable role for FYN-TRAF3IP2 in PTCLs and call for the clinical testing of anti-NF-κB targeted therapies in these diseases.


Subject(s)
Immunoblastic Lymphadenopathy , Lymphoma, T-Cell, Peripheral , Adaptor Proteins, Signal Transducing/genetics , Animals , Immunoblastic Lymphadenopathy/genetics , Lymphoma, T-Cell, Peripheral/genetics , Mice , NF-kappa B/genetics , Oncogenes , Signal Transduction
8.
Article in English | MEDLINE | ID: mdl-32513675

ABSTRACT

Peripheral T-cell lymphomas (PTCLs) constitute a highly heterogeneous group of hematological diseases with complex clinical and molecular features consistent with the diversity of the T-cell type from which they originate. In the past several years, the systematic implementation of high-throughput genomic technologies for the analysis of T-cell malignancies has supported an exponential progress in our understanding of the genetic drivers of oncogenesis and unraveled the molecular complexity of these diseases. Recent findings have helped redefine the classification of T-cell malignancies and provided novel biomarkers to improve diagnosis accuracy and analyze the response to therapy. In addition, multiple novel targeted therapies including small-molecule inhibitors, antibody-based approaches, and immunotherapy have shown promising results in early clinical analysis and have the potential to completely change the way T-cell malignancies have been treated traditionally.


Subject(s)
Cell Transformation, Neoplastic/pathology , Lymphoma, T-Cell, Peripheral/pathology , Biomarkers , Humans , Lymphoma, T-Cell, Peripheral/genetics
9.
Am J Hematol ; 94(6): 628-634, 2019 06.
Article in English | MEDLINE | ID: mdl-30829413

ABSTRACT

The histological diagnosis of peripheral T-cell lymphoma (PTCL) can represent a challenge, particularly in the case of closely related entities such as angioimmunoblastic T-lymphoma (AITL), PTCL-not otherwise specified (PTCL-NOS), and ALK-negative anaplastic large-cell lymphoma (ALCL). Although gene expression profiling and next generations sequencing have been proven to define specific features recurrently associated with distinct entities, genomic-based stratifications have not yet led to definitive diagnostic criteria and/or entered into the routine clinical practice. Herein, to improve the current molecular classification between AITL and PTCL-NOS, we analyzed the transcriptional profiles from 503 PTCLs stratified according to their molecular configuration and integrated them with genomic data of recurrently mutated genes (RHOA G17V , TET2, IDH2 R172 , and DNMT3A) in 53 cases (39 AITLs and 14 PTCL-NOSs) included in the series. Our analysis unraveled that the mutational status of RHOA G17V , TET2, and DNMT3A poorly correlated, individually, with peculiar transcriptional fingerprints. Conversely, in IDH2 R172 samples a strong transcriptional signature was identified that could act as a surrogate for mutational status. The integrated analysis of clinical, mutational, and molecular data led to a simplified 19-gene signature that retains high accuracy in differentiating the main nodal PTCL entities. The expression levels of those genes were confirmed in an independent cohort profiled by RNA-sequencing.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Lymphoma, T-Cell, Peripheral , Mutation , Neoplasm Proteins , Transcription, Genetic , Female , Humans , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/metabolism , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics
10.
Cancer Cell ; 33(2): 259-273.e7, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29398449

ABSTRACT

Angioimmunoblastic T cell lymphoma (AITL) is an aggressive tumor derived from malignant transformation of T follicular helper (Tfh) cells. AITL is characterized by loss-of-function mutations in Ten-Eleven Translocation 2 (TET2) epigenetic tumor suppressor and a highly recurrent mutation (p.Gly17Val) in the RHOA small GTPase. Yet, the specific role of RHOA G17V in AITL remains unknown. Expression of Rhoa G17V in CD4+ T cells induces Tfh cell specification; increased proliferation associated with inducible co-stimulator (ICOS) upregulation and increased phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase signaling. Moreover, RHOA G17V expression together with Tet2 loss resulted in development of AITL in mice. Importantly, Tet2-/-RHOA G17V tumor proliferation in vivo can be inhibited by ICOS/PI3K-specific blockade, supporting a driving role for ICOS signaling in Tfh cell transformation.


Subject(s)
DNA-Binding Proteins/genetics , Immunoblastic Lymphadenopathy/genetics , Mutation/genetics , Proto-Oncogene Proteins/genetics , T-Lymphocytes, Helper-Inducer/immunology , rhoA GTP-Binding Protein/metabolism , Animals , Biomarkers, Tumor/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Lymphoma, T-Cell/metabolism , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 114(4): 764-769, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28062691

ABSTRACT

Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of non-Hodgkin lymphomas frequently associated with poor prognosis and for which genetic mechanisms of transformation remain incompletely understood. Using RNA sequencing and targeted sequencing, here we identify a recurrent in-frame deletion (VAV1 Δ778-786) generated by a focal deletion-driven alternative splicing mechanism as well as novel VAV1 gene fusions (VAV1-THAP4, VAV1-MYO1F, and VAV1-S100A7) in PTCL. Mechanistically these genetic lesions result in increased activation of VAV1 catalytic-dependent (MAPK, JNK) and non-catalytic-dependent (nuclear factor of activated T cells, NFAT) VAV1 effector pathways. These results support a driver oncogenic role for VAV1 signaling in the pathogenesis of PTCL.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Guanine/metabolism , Lymphoma, T-Cell, Peripheral/genetics , Mutation/genetics , Proto-Oncogene Proteins c-vav/genetics , Translocation, Genetic/genetics , Alternative Splicing/genetics , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Humans , Jurkat Cells , Sequence Deletion/genetics
13.
Proc Natl Acad Sci U S A ; 113(40): 11306-11311, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27655895

ABSTRACT

Although multiagent combination chemotherapy is curative in a significant fraction of childhood acute lymphoblastic leukemia (ALL) patients, 20% of cases relapse and most die because of chemorefractory disease. Here we used whole-exome and whole-genome sequencing to analyze the mutational landscape at relapse in pediatric ALL cases. These analyses identified numerous relapse-associated mutated genes intertwined in chemotherapy resistance-related protein complexes. In this context, RAS-MAPK pathway-activating mutations in the neuroblastoma RAS viral oncogene homolog (NRAS), kirsten rat sarcoma viral oncogene homolog (KRAS), and protein tyrosine phosphatase, nonreceptor type 11 (PTPN11) genes were present in 24 of 55 (44%) cases in our series. Interestingly, some leukemias showed retention or emergence of RAS mutant clones at relapse, whereas in others RAS mutant clones present at diagnosis were replaced by RAS wild-type populations, supporting a role for both positive and negative selection evolutionary pressures in clonal evolution of RAS-mutant leukemia. Consistently, functional dissection of mouse and human wild-type and mutant RAS isogenic leukemia cells demonstrated induction of methotrexate resistance but also improved the response to vincristine in mutant RAS-expressing lymphoblasts. These results highlight the central role of chemotherapy-driven selection as a central mechanism of leukemia clonal evolution in relapsed ALL, and demonstrate a previously unrecognized dual role of RAS mutations as drivers of both sensitivity and resistance to chemotherapy.


Subject(s)
Clonal Evolution/genetics , Genes, ras , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Base Sequence , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Methotrexate/pharmacology , Methotrexate/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Vincristine/pharmacology , Vincristine/therapeutic use
14.
Curr Opin Hematol ; 23(4): 434-43, 2016 07.
Article in English | MEDLINE | ID: mdl-27177312

ABSTRACT

PURPOSE OF REVIEW: Once an obscure disease, recent studies have transformed our understanding of angioimmunoblastic T-cell lymphoma (AITL). In this review, we summarize new major advances in the genetics and biology of AITL. RECENT FINDINGS: Genome wide sequencing studies have dissected the repertoire of the genetic alterations driving AITL uncovering a highly recurrent Gly17Val somatic mutation in the small GTPase RHOA and major role for mutations in epigenetic regulators, such as TET2, DNMT3A and IDH2, and signaling factors (e.g., FYN and CD28). These findings support a multistep model of follicular T helper cell transformation in AITL and pinpoint novel candidates for the development of targeted therapies in this disease. SUMMARY: AITL originates from follicular T helper cells and is characterized by the presence of RHOA G17V mutation together with genetic alterations in TET2, DNMT3A, and IDH2. Research efforts now focus on the elucidation of the specific roles and interplay of these genetic alterations in the pathogenesis of AITL.


Subject(s)
Immunoblastic Lymphadenopathy/etiology , Lymphoma, T-Cell/etiology , Animals , Biomarkers, Tumor , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genomics/methods , Humans , Immunoblastic Lymphadenopathy/diagnosis , Immunoblastic Lymphadenopathy/metabolism , Lymphoma, T-Cell/diagnosis , Lymphoma, T-Cell/metabolism , Mutation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology , Transcriptome , rhoA GTP-Binding Protein/genetics
15.
Pediatr Blood Cancer ; 63(9): 1660-3, 2016 09.
Article in English | MEDLINE | ID: mdl-27149388

ABSTRACT

A 4-year-old male with the diagnosis of T-cell acute lymphoblastic leukemia (T-ALL) relapsed after 19 months with an acute myeloid leukemia (AML). Immunoglobulin and T-cell receptor gene rearrangements analyses reveal that both leukemias were rearranged with a clonal relationship between them. Comparative genomic hybridization (Array-CGH) and whole-exome sequencing analyses of both samples suggest that this leukemia may have originated from a common T/myeloid progenitor. The presence of homozygous deletion of p16/INK4A, p14/ARF, p15/INK4B, and heterozygous deletion of WT1 locus remained stable in the leukemia throughout phenotypic switch, revealing that this AML can be genetically associated to T-ALL.


Subject(s)
Leukemia, Myeloid, Acute/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/complications , Child, Preschool , Gene Deletion , Gene Rearrangement, T-Lymphocyte , Humans , Leukemia, Myeloid, Acute/genetics , Male , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Recurrence
16.
Nat Genet ; 47(12): 1465-70, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26551667

ABSTRACT

Sézary syndrome is a leukemic and aggressive form of cutaneous T cell lymphoma (CTCL) resulting from the malignant transformation of skin-homing central memory CD4(+) T cells. Here we performed whole-exome sequencing of tumor-normal sample pairs from 25 patients with Sézary syndrome and 17 patients with other CTCLs. These analyses identified a distinctive pattern of somatic copy number alterations in Sézary syndrome, including highly prevalent chromosomal deletions involving the TP53, RB1, PTEN, DNMT3A and CDKN1B tumor suppressors. Mutation analysis identified a broad spectrum of somatic mutations in key genes involved in epigenetic regulation (TET2, CREBBP, KMT2D (MLL2), KMT2C (MLL3), BRD9, SMARCA4 and CHD3) and signaling, including MAPK1, BRAF, CARD11 and PRKG1 mutations driving increased MAPK, NF-κB and NFAT activity upon T cell receptor stimulation. Collectively, our findings provide new insights into the genetics of Sézary syndrome and CTCL and support the development of personalized therapies targeting key oncogenically activated signaling pathways for the treatment of these diseases.


Subject(s)
Gene Expression Regulation, Neoplastic , Genetic Markers/genetics , Lymphoma, T-Cell, Cutaneous/genetics , Mutation/genetics , Sezary Syndrome/genetics , Skin Neoplasms/genetics , Case-Control Studies , Chromosome Deletion , DNA Mutational Analysis , Epigenesis, Genetic , Exome/genetics , Genetic Loci , Humans , Immunoblotting , Lymphoma, T-Cell, Cutaneous/metabolism , Lymphoma, T-Cell, Cutaneous/pathology , Risk Factors , Sezary Syndrome/metabolism , Sezary Syndrome/pathology , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
17.
Nat Med ; 20(10): 1130-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25194570

ABSTRACT

Efforts to identify and annotate cancer driver genetic lesions have been focused primarily on the analysis of protein-coding genes; however, most genetic abnormalities found in human cancer are located in intergenic regions. Here we identify a new long range-acting MYC enhancer controlled by NOTCH1 that is targeted by recurrent chromosomal duplications in human T cell acute lymphoblastic leukemia (T-ALL). This highly conserved regulatory element, hereby named N-Me for NOTCH MYC enhancer, is located within a broad super-enhancer region +1.47 Mb from the MYC transcription initiating site, interacts with the MYC proximal promoter and induces orientation-independent MYC expression in reporter assays. Moreover, analysis of N-Me knockout mice demonstrates a selective and essential role of this regulatory element during thymocyte development and in NOTCH1-induced T-ALL. Together these results identify N-Me as a long-range oncogenic enhancer implicated directly in the pathogenesis of human leukemia and highlight the importance of the NOTCH1-MYC regulatory axis in T cell transformation and as a therapeutic target in T-ALL.


Subject(s)
Enhancer Elements, Genetic , Genes, myc , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptor, Notch1/genetics , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Female , Gene Amplification , Humans , Jurkat Cells , Lymphopoiesis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oncogenes , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
18.
Nat Genet ; 46(2): 166-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24413734

ABSTRACT

Peripheral T cell lymphomas (PTCLs) are a heterogeneous and poorly understood group of non-Hodgkin lymphomas. Here we combined whole-exome sequencing of 12 tumor-normal DNA pairs, RNA sequencing analysis and targeted deep sequencing to identify new genetic alterations in PTCL transformation. These analyses identified highly recurrent epigenetic factor mutations in TET2, DNMT3A and IDH2 as well as a new highly prevalent RHOA mutation encoding a p.Gly17Val alteration present in 22 of 35 (67%) angioimmunoblastic T cell lymphoma (AITL) samples and in 8 of 44 (18%) PTCL, not otherwise specified (PTCL-NOS) samples. Mechanistically, the RHOA Gly17Val protein interferes with RHOA signaling in biochemical and cellular assays, an effect potentially mediated by the sequestration of activated guanine-exchange factor (GEF) proteins. In addition, we describe new and recurrent, albeit less frequent, genetic defects including mutations in FYN, ATM, B2M and CD58 implicating SRC signaling, impaired DNA damage response and escape from immune surveillance mechanisms in the pathogenesis of PTCL.


Subject(s)
Epigenesis, Genetic/genetics , Lymphoma, T-Cell, Peripheral/genetics , Proto-Oncogene Proteins c-fyn/genetics , rhoA GTP-Binding Protein/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Base Sequence , CD58 Antigens/genetics , Computational Biology , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , Dioxygenases , Escherichia coli , Exome/genetics , Fluorescent Antibody Technique , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Isocitrate Dehydrogenase/genetics , Molecular Sequence Data , Mutation, Missense/genetics , Proto-Oncogene Proteins/genetics , Sequence Analysis, RNA
19.
Nat Med ; 19(3): 368-71, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23377281

ABSTRACT

Acute lymphoblastic leukemia (ALL) is an aggressive hematological tumor resulting from the malignant transformation of lymphoid progenitors. Despite intensive chemotherapy, 20% of pediatric patients and over 50% of adult patients with ALL do not achieve a complete remission or relapse after intensified chemotherapy, making disease relapse and resistance to therapy the most substantial challenge in the treatment of this disease. Using whole-exome sequencing, we identify mutations in the cytosolic 5'-nucleotidase II gene (NT5C2), which encodes a 5'-nucleotidase enzyme that is responsible for the inactivation of nucleoside-analog chemotherapy drugs, in 20/103 (19%) relapse T cell ALLs and 1/35 (3%) relapse B-precursor ALLs. NT5C2 mutant proteins show increased nucleotidase activity in vitro and conferred resistance to chemotherapy with 6-mercaptopurine and 6-thioguanine when expressed in ALL lymphoblasts. These results support a prominent role for activating mutations in NT5C2 and increased nucleoside-analog metabolism in disease progression and chemotherapy resistance in ALL.


Subject(s)
5'-Nucleotidase/genetics , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Mercaptopurine/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , 5'-Nucleotidase/metabolism , Arabinonucleosides/pharmacology , Arabinonucleosides/therapeutic use , Base Sequence , Cell Line , HEK293 Cells , Humans , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Recurrence , Sequence Analysis, DNA , Thioguanine/therapeutic use
20.
Cancer Cell ; 22(2): 222-34, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22897852

ABSTRACT

Here, we have investigated the role of the Notch pathway in the generation and maintenance of Kras(G12V)-driven non-small cell lung carcinomas (NSCLCs). We demonstrate by genetic means that γ-secretase and RBPJ are essential for the formation of NSCLCs. Of importance, pharmacologic treatment of mice carrying autochthonous NSCLCs with a γ-secretase inhibitor (GSI) blocks cancer growth. Treated carcinomas present reduced HES1 levels and reduced phosphorylated ERK without changes in phosphorylated MEK. Mechanistically, we show that HES1 directly binds to and represses the promoter of DUSP1, encoding a dual phosphatase that is active against phospho-ERK. Accordingly, GSI treatment upregulates DUSP1 and decreases phospho-ERK. These data provide proof of the in vivo therapeutic potential of GSIs in primary NSCLCs.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Enzyme Inhibitors/therapeutic use , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Lung Neoplasms/drug therapy , ras Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Dual Specificity Phosphatase 1/metabolism , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Homeodomain Proteins/metabolism , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mutant Proteins/metabolism , Phosphorylation/drug effects , Presenilin-1/metabolism , Presenilin-2/metabolism , Prognosis , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Signal Transduction/drug effects , Transcription Factor HES-1 , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...