201
|
PTEN microdeletions in T-cell acute lymphoblastic leukemia are caused by illegitimate RAG-mediated recombination events. Blood 2014; 124:567-78. [PMID: 24904117 DOI: 10.1182/blood-2014-03-562751] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Phosphatase and tensin homolog (PTEN)-inactivating mutations and/or deletions are an independent risk factor for relapse of T-cell acute lymphoblastic leukemia (T-ALL) patients treated on Dutch Childhood Oncology Group or German Cooperative Study Group for Childhood Acute Lymphoblastic Leukemia protocols. Some monoallelic mutated or PTEN wild-type patients lack PTEN protein, implying that additional PTEN inactivation mechanisms exist. We show that PTEN is inactivated by small deletions affecting a few exons in 8% of pediatric T-ALL patients. These microdeletions were clonal in 3% and subclonal in 5% of patients. Conserved deletion breakpoints are flanked by cryptic recombination signal sequences (cRSSs) and frequently have non-template-derived nucleotides inserted in between breakpoints, pointing to an illegitimate RAG recombination-driven activity. Identified cRSSs drive RAG-dependent recombination in a reporter system as efficiently as bona fide RSSs that flank gene segments of the T-cell receptor locus. Remarkably, equivalent microdeletions were detected in thymocytes of healthy individuals. Microdeletions strongly associate with the TALLMO subtype characterized by TAL1 or LMO2 rearrangements. Primary and secondary xenotransplantation of TAL1-rearranged leukemia allowed development of leukemic subclones with newly acquired PTEN microdeletions. Ongoing RAG activity may therefore actively contribute to the acquisition of preleukemic hits, clonal diversification, and disease progression.
Collapse
|
202
|
Uzan B, Poglio S, Gerby B, Wu CL, Gross J, Armstrong F, Calvo J, Cahu X, Deswarte C, Dumont F, Passaro D, Besnard-Guérin C, Leblanc T, Baruchel A, Landman-Parker J, Ballerini P, Baud V, Ghysdael J, Baleydier F, Porteu F, Pflumio F. Interleukin-18 produced by bone marrow-derived stromal cells supports T-cell acute leukaemia progression. EMBO Mol Med 2014; 6:821-34. [PMID: 24778454 PMCID: PMC4203358 DOI: 10.1002/emmm.201303286] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Development of novel therapies is critical for T-cell acute leukaemia (T-ALL). Here, we investigated the effect of inhibiting the MAPK/MEK/ERK pathway on T-ALL cell growth. Unexpectedly, MEK inhibitors (MEKi) enhanced growth of 70% of human T-ALL cell samples cultured on stromal cells independently of NOTCH activation and maintained their ability to propagate in vivo. Similar results were obtained when T-ALL cells were cultured with ERK1/2-knockdown stromal cells or with conditioned medium from MEKi-treated stromal cells. Microarray analysis identified interleukin 18 (IL-18) as transcriptionally up-regulated in MEKi-treated MS5 cells. Recombinant IL-18 promoted T-ALL growth in vitro, whereas the loss of function of IL-18 receptor in T-ALL blast cells decreased blast proliferation in vitro and in NSG mice. The NFKB pathway that is downstream to IL-18R was activated by IL-18 in blast cells. IL-18 circulating levels were increased in T-ALL-xenografted mice and also in T-ALL patients in comparison with controls. This study uncovers a novel role of the pro-inflammatory cytokine IL-18 and outlines the microenvironment involvement in human T-ALL development.
Collapse
Affiliation(s)
- Benjamin Uzan
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL Equipe Labellisée Ligue Contre le Cancer UMR 967, Fontenay-aux-Roses, France INSERM U967, Fontenay-aux-Roses, France Université Paris Diderot Sorbonne Paris Cité UMR 967, Fontenay-aux-Roses, France Université Paris-Sud UMR 967, Fontenay-aux-Roses, France
| | - Sandrine Poglio
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL Equipe Labellisée Ligue Contre le Cancer UMR 967, Fontenay-aux-Roses, France INSERM U967, Fontenay-aux-Roses, France Université Paris Diderot Sorbonne Paris Cité UMR 967, Fontenay-aux-Roses, France Université Paris-Sud UMR 967, Fontenay-aux-Roses, France
| | - Bastien Gerby
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL Equipe Labellisée Ligue Contre le Cancer UMR 967, Fontenay-aux-Roses, France INSERM U967, Fontenay-aux-Roses, France Université Paris Diderot Sorbonne Paris Cité UMR 967, Fontenay-aux-Roses, France Université Paris-Sud UMR 967, Fontenay-aux-Roses, France
| | - Ching-Lien Wu
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL Equipe Labellisée Ligue Contre le Cancer UMR 967, Fontenay-aux-Roses, France INSERM U967, Fontenay-aux-Roses, France Université Paris Diderot Sorbonne Paris Cité UMR 967, Fontenay-aux-Roses, France Université Paris-Sud UMR 967, Fontenay-aux-Roses, France
| | - Julia Gross
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL Equipe Labellisée Ligue Contre le Cancer UMR 967, Fontenay-aux-Roses, France INSERM U967, Fontenay-aux-Roses, France Université Paris Diderot Sorbonne Paris Cité UMR 967, Fontenay-aux-Roses, France Université Paris-Sud UMR 967, Fontenay-aux-Roses, France
| | - Florence Armstrong
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL Equipe Labellisée Ligue Contre le Cancer UMR 967, Fontenay-aux-Roses, France INSERM U967, Fontenay-aux-Roses, France Université Paris Diderot Sorbonne Paris Cité UMR 967, Fontenay-aux-Roses, France Université Paris-Sud UMR 967, Fontenay-aux-Roses, France
| | - Julien Calvo
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL Equipe Labellisée Ligue Contre le Cancer UMR 967, Fontenay-aux-Roses, France INSERM U967, Fontenay-aux-Roses, France Université Paris Diderot Sorbonne Paris Cité UMR 967, Fontenay-aux-Roses, France Université Paris-Sud UMR 967, Fontenay-aux-Roses, France
| | - Xavier Cahu
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL Equipe Labellisée Ligue Contre le Cancer UMR 967, Fontenay-aux-Roses, France INSERM U967, Fontenay-aux-Roses, France Université Paris Diderot Sorbonne Paris Cité UMR 967, Fontenay-aux-Roses, France Université Paris-Sud UMR 967, Fontenay-aux-Roses, France
| | - Caroline Deswarte
- Service D'hématologie Pédiatrique, Assistance Publique - Hôpitaux de Paris Hôpital A. Trousseau, Paris, France
| | - Florent Dumont
- INSERM U1016 Institut Cochin, Paris, France CNRS UMR8104, Paris, France Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Diana Passaro
- Institut Curie Centre Universitaire, Orsay, France CNRS UMR 3306, Orsay, France Institut National de la Santé et de la Recherche Médicale U1005, Orsay, France
| | - Corinne Besnard-Guérin
- INSERM U1016 Institut Cochin, Paris, France CNRS UMR8104, Paris, France Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Thierry Leblanc
- Service D'hématologie Pédiatrique, Assistance Publique - Hôpitaux de Paris Hôpital Robert Debré, Paris, France
| | - André Baruchel
- Service D'hématologie Pédiatrique, Assistance Publique - Hôpitaux de Paris Hôpital Robert Debré, Paris, France
| | - Judith Landman-Parker
- Service D'hématologie Pédiatrique, Assistance Publique - Hôpitaux de Paris Hôpital A. Trousseau, Paris, France
| | - Paola Ballerini
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL Equipe Labellisée Ligue Contre le Cancer UMR 967, Fontenay-aux-Roses, France INSERM U967, Fontenay-aux-Roses, France Université Paris Diderot Sorbonne Paris Cité UMR 967, Fontenay-aux-Roses, France Université Paris-Sud UMR 967, Fontenay-aux-Roses, France Service D'hématologie Pédiatrique, Assistance Publique - Hôpitaux de Paris Hôpital A. Trousseau, Paris, France
| | - Véronique Baud
- INSERM U1016 Institut Cochin, Paris, France CNRS UMR8104, Paris, France Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Jacques Ghysdael
- Institut Curie Centre Universitaire, Orsay, France CNRS UMR 3306, Orsay, France Institut National de la Santé et de la Recherche Médicale U1005, Orsay, France
| | - Frédéric Baleydier
- Institut d'Hématologie et Oncologie Pédiatrique Hospices Civils de Lyon et Université Claude Bernard, Lyon, France
| | - Francoise Porteu
- INSERM U1016 Institut Cochin, Paris, France CNRS UMR8104, Paris, France Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Francoise Pflumio
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA) DSV-IRCM-SCSR-LSHL Equipe Labellisée Ligue Contre le Cancer UMR 967, Fontenay-aux-Roses, France INSERM U967, Fontenay-aux-Roses, France Université Paris Diderot Sorbonne Paris Cité UMR 967, Fontenay-aux-Roses, France Université Paris-Sud UMR 967, Fontenay-aux-Roses, France
| |
Collapse
|
203
|
Treanor LM, Zhou S, Janke L, Churchman ML, Ma Z, Lu T, Chen SC, Mullighan CG, Sorrentino BP. Interleukin-7 receptor mutants initiate early T cell precursor leukemia in murine thymocyte progenitors with multipotent potential. ACTA ACUST UNITED AC 2014; 211:701-13. [PMID: 24687960 PMCID: PMC3978278 DOI: 10.1084/jem.20122727] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Early T cell precursor acute lymphoblastic leukemia (ETP-ALL) exhibits lymphoid, myeloid, and stem cell features and is associated with a poor prognosis. Whole genome sequencing of human ETP-ALL cases has identified recurrent mutations in signaling, histone modification, and hematopoietic development genes but it remains to be determined which of these abnormalities are sufficient to initiate leukemia. We show that activating mutations in the interleukin-7 receptor identified in human pediatric ETP-ALL cases are sufficient to generate ETP-ALL in mice transplanted with primitive transduced thymocytes from p19(Arf-/-) mice. The cellular mechanism by which these mutant receptors induce ETP-ALL is the block of thymocyte differentiation at the double negative 2 stage at which myeloid lineage and T lymphocyte developmental potential coexist. Analyses of samples from pediatric ETP-ALL cases and our murine ETP-ALL model show uniformly high levels of LMO2 expression, very low to undetectable levels of BCL11B expression, and a relative lack of activating NOTCH1 mutations. We report that pharmacological blockade of Jak-Stat signaling with ruxolitinib has significant antileukemic activity in this ETP-ALL model. This new murine model recapitulates several important cellular and molecular features of ETP-ALL and should be useful to further define novel therapeutic approaches for this aggressive leukemia.
Collapse
Affiliation(s)
- Louise M Treanor
- Department of Hematology and 2 Department of Pathology, Division of Experimental Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Rozovski U, Li P, Harris D, Ohanian M, Kantarjian H, Estrov Z. Interleukin-7 receptor-α gene mutations are not detected in adult T-cell acute lymphoblastic leukemia. Cancer Med 2014; 3:550-4. [PMID: 24678068 PMCID: PMC4101745 DOI: 10.1002/cam4.194] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/23/2013] [Accepted: 12/26/2013] [Indexed: 12/21/2022] Open
Abstract
Somatic mutations in cancer cell genes are classified according to their functional significance. Those that provide the malignant cells with significant advantage are collectively referred to as driver mutations and those that do not, are the passenger mutations. Accordingly, analytical criteria to distinguish driver mutations from passenger mutations have been recently suggested. Recent studies revealed mutations in interleukin-7 receptor-α (IL7R) gene in 10% of pediatric T-cell acute lymphoblastic leukemia (T-ALL) patients and in only a few cases of pediatric B-ALL. IL7R mutations are also frequently found in patients with lung cancer, but whereas in pediatric T-ALL IL7R mutations are “drivers” (consisting of gain-of-function mutations within a narrow 50-base pair interval at exon 6 that confer cytokine-independent cell growth and promote tumor transformation), in lung cancer, mutations are substitution mutations randomly distributed across the gene and are probably only “passenger” events. Because the treatment response of adult T-ALL is significantly poorer than that of childhood T-ALL and because exon 6 IL7R mutations play a role in the pathogenesis of childhood T-ALL, we sought to determine how the pattern of IL7R mutations varies between adult and childhood T-ALL. To that end, we sequenced the 50-base pair interval in exon 6 of the IL7R of DNA obtained from bone marrow samples of 35 randomly selected adult patients with T-ALL. Our analysis revealed that none of these 35 samples carried an IL7R mutation in exon 6. Whether differences in the genetic makeup of adult and childhood T-ALL explain the differential response to therapy remains to be determined.
Collapse
Affiliation(s)
- Uri Rozovski
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | |
Collapse
|
205
|
Verstraete K, van Schie L, Vyncke L, Bloch Y, Tavernier J, Pauwels E, Peelman F, Savvides SN. Structural basis of the proinflammatory signaling complex mediated by TSLP. Nat Struct Mol Biol 2014; 21:375-82. [PMID: 24632570 DOI: 10.1038/nsmb.2794] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 02/18/2014] [Indexed: 02/08/2023]
Abstract
Thymic stromal lymphopoietin (TSLP), a cytokine produced by epithelial cells at barrier surfaces, is pivotal for the development of widespread chronic inflammatory disorders such as asthma and atopic dermatitis. The structure of the mouse TSLP-mediated signaling complex reveals how TSLP establishes extensive interfaces with its cognate receptor (TSLPR) and the shared interleukin 7 receptor α-chain (IL-7Rα) to evoke membrane-proximal receptor-receptor contacts poised for intracellular signaling. Binding of TSLP to TSLPR is a mechanistic prerequisite for recruitment of IL-7Rα to the high-affinity ternary complex, which we propose is coupled to a structural switch in TSLP at the crossroads of the cytokine-receptor interfaces. Functional interrogation of TSLP-receptor interfaces points to putative interaction hotspots that could be exploited for antagonist design. Finally, we derive the structural rationale for the functional duality of IL-7Rα and establish a consensus for the geometry of ternary complexes mediated by interleukin 2 (IL-2)-family cytokines.
Collapse
Affiliation(s)
- Kenneth Verstraete
- Unit for Structural Biology, Laboratory for Protein Biochemistry and Biomolecular Engineering, Department of Biochemistry & Microbiology, Ghent University, Ghent, Belgium
| | - Loes van Schie
- Unit for Structural Biology, Laboratory for Protein Biochemistry and Biomolecular Engineering, Department of Biochemistry & Microbiology, Ghent University, Ghent, Belgium
| | - Laurens Vyncke
- Department of Medical Protein Research, Vlaams Interuniversitair Instituut voor Biotechnologie and Ghent University, Ghent, Belgium
| | - Yehudi Bloch
- Unit for Structural Biology, Laboratory for Protein Biochemistry and Biomolecular Engineering, Department of Biochemistry & Microbiology, Ghent University, Ghent, Belgium
| | - Jan Tavernier
- Department of Medical Protein Research, Vlaams Interuniversitair Instituut voor Biotechnologie and Ghent University, Ghent, Belgium
| | - Ewald Pauwels
- Center for Molecular Modeling, Ghent University, Ghent, Belgium
| | - Frank Peelman
- Department of Medical Protein Research, Vlaams Interuniversitair Instituut voor Biotechnologie and Ghent University, Ghent, Belgium
| | - Savvas N Savvides
- Unit for Structural Biology, Laboratory for Protein Biochemistry and Biomolecular Engineering, Department of Biochemistry & Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
206
|
Abstract
In the current issue of Blood, Yokoyama et al demonstrate that an IL7R mutation similar to those found in patients with acute lymphoblastic leukemia (ALL) can be leukemogenic in vivo when expressed in normal hematopoietic progenitors.
Collapse
|
207
|
Izraeli S, Shochat C, Tal N, Geron I. Towards precision medicine in childhood leukemia--insights from mutationally activated cytokine receptor pathways in acute lymphoblastic leukemia. Cancer Lett 2014; 352:15-20. [PMID: 24569093 DOI: 10.1016/j.canlet.2014.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 02/08/2014] [Accepted: 02/10/2014] [Indexed: 01/30/2023]
Abstract
The successful therapy of childhood leukemia has been characterized by careful personalized adaptation of therapy by risk stratification. Yet almost all drugs are relatively non-specific. To achieve greater precision in therapy, druggable targets and specific targeting drugs are necessary. Here we review the recent discoveries of cytokine receptors and their signaling components in high risk leukemias and the potential approaches to target them.
Collapse
Affiliation(s)
- Shai Izraeli
- Childhood Leukemia Research Section, Department of Pediatric Hemato-Oncology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel.
| | - Chen Shochat
- Childhood Leukemia Research Section, Department of Pediatric Hemato-Oncology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel; Migal Galilee Technology Center, Kiryat Shmona, Israel; Tel Hai College, Upper Galilee 12210, Israel
| | - Noa Tal
- Childhood Leukemia Research Section, Department of Pediatric Hemato-Oncology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Ifat Geron
- Childhood Leukemia Research Section, Department of Pediatric Hemato-Oncology, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel; Division of Biological Sciences and Department of Medicine Stem Cell Program, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
208
|
Mughal TI, Girnius S, Rosen ST, Kumar S, Wiestner A, Abdel-Wahab O, Kiladjian JJ, Wilson WH, Van Etten RA. Emerging therapeutic paradigms to target the dysregulated Janus kinase/signal transducer and activator of transcription pathway in hematological malignancies. Leuk Lymphoma 2014; 55:1968-79. [PMID: 24206094 DOI: 10.3109/10428194.2013.863307] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Over the past decade, there has been increasing biochemical evidence that the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway is aberrantly activated in malignant cells from patients with a wide spectrum of cancers of the blood and immune systems. The emerging availability of small molecule inhibitors of JAK and other signaling molecules in the JAK/STAT pathway has allowed preclinical studies validating an important role of this pathway in the pathogenesis of many hematologic malignancies, and provided motivation for new strategies for treatment of these diseases. Here, a round-table panel of experts review the current preclinical and clinical landscape of the JAK/STAT pathway in acute lymphoid and myeloid leukemias, lymphomas and myeloma, and chronic myeloid neoplasms.
Collapse
|
209
|
Tal N, Shochat C, Geron I, Bercovich D, Izraeli S. Interleukin 7 and thymic stromal lymphopoietin: from immunity to leukemia. Cell Mol Life Sci 2014; 71:365-78. [PMID: 23625073 PMCID: PMC11113825 DOI: 10.1007/s00018-013-1337-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/10/2013] [Accepted: 04/08/2013] [Indexed: 01/12/2023]
Abstract
Cancer is often caused by deregulation of normal developmental processes. Here, we review recent research on the aberrant activation of two hematopoietic cytokine receptors in acute lymphoid leukemias. Somatic events in the genes for thymic stromal lymphopoietin and Interleukin 7 receptors as well as in their downstream JAK kinases result in constitutive ligand-independent activation of survival and proliferation in B and T lymphoid precursors. Drugs targeting these receptors or the signaling pathways might provide effective therapies of these leukemias.
Collapse
Affiliation(s)
- Noa Tal
- Cancer Research Center, Sheba Medical Center, Edmond and Lily Safra Children’s Hospital, Tel Hashomer, 52621 Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chen Shochat
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Migal Galilee Technology Center, Kiryat Shmona, Israel
- Tel Hai College, 12210 Upper Galilee, Israel
| | - Ifat Geron
- Cancer Research Center, Sheba Medical Center, Edmond and Lily Safra Children’s Hospital, Tel Hashomer, 52621 Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Biological Sciences and Department of Medicine Stem Cell Program, University of California San Diego, La Jolla, California USA
| | - Dani Bercovich
- Migal Galilee Technology Center, Kiryat Shmona, Israel
- Tel Hai College, 12210 Upper Galilee, Israel
| | - Shai Izraeli
- Cancer Research Center, Sheba Medical Center, Edmond and Lily Safra Children’s Hospital, Tel Hashomer, 52621 Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
210
|
Abstract
T-cell neoplasms include both mature T-cell leukemias and lymphomas and immature proliferations of precursor T cells. Molecular laboratories routinely assay suspected T-cell proliferations for evidence of clonality. In addition, some T-cell neoplasms are characterized by recurrent structural abnormalities that can be readily identified by such techniques as fluorescence in situ hybridization. New massively parallel sequencing technologies have led to the identification of numerous recurrent gene mutations in T-cell neoplasms. These findings are reviewed. As new technologies become implemented in molecular diagnostic laboratories and as targeted therapies are developed, it is anticipated that more extensive genomic characterization of T-cell neoplasms will be routinely performed in the future.
Collapse
|
211
|
Abstract
Abstract
Children with Down syndrome are at high risk for developing B-cell precursor acute lymphoblastic leukemia (DS-ALL) associated with poor outcome due to both a high relapse rate and increased treatment-related mortality (TRM) from infections. Biologically, these heterogeneous leukemias are characterized by under-representation of the common cytogenetic subgroups of childhood ALL and overrepresentation of CRLF2-IL7R-JAK-STAT activating genetic aberrations. Although relapse is the major determinant of poor outcomes in this population, de-escalation of chemotherapy intensity might be feasible in the 10% to 15% DS-ALL patients with ETV6-RUNX1 or high hyperdipoidy in whom TRM is the major limiting event. As infection-associated TRM occurs during all treatment phases, including the maintenance period, increased surveillance and supportive care is required throughout therapy. Improvement in outcome will require better understanding of the causes of treatment failure and TRM, incorporation of new therapies targeting the unique biological properties of DS-ALL, and enhanced supportive care measures to reduce the risk of infection-related TRM. To facilitate these goals, an international collaboration plans to establish a prospective DS-ALL registry and develop specific supportive care recommendations for this at-risk population.
Collapse
|
212
|
Hales EC, Taub JW, Matherly LH. New insights into Notch1 regulation of the PI3K–AKT–mTOR1 signaling axis: Targeted therapy of γ-secretase inhibitor resistant T-cell acute lymphoblastic leukemia. Cell Signal 2014; 26:149-61. [DOI: 10.1016/j.cellsig.2013.09.021] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 09/30/2013] [Indexed: 02/01/2023]
|
213
|
JAK2 and myeloproliferative neoplasms. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
214
|
In vivo leukemogenic potential of an interleukin 7 receptor α chain mutant in hematopoietic stem and progenitor cells. Blood 2013; 122:4259-63. [DOI: 10.1182/blood-2012-08-451278] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Key Points
Gain-of function mutation of IL7Rα induces lymphoid leukemia as well as myeloproliferative disease. In vivo oncogenicity of mutant IL7Rα is influenced by the differentiation stage at which it occurs.
Collapse
|
215
|
Kalender Atak Z, Gianfelici V, Hulselmans G, De Keersmaecker K, Devasia AG, Geerdens E, Mentens N, Chiaretti S, Durinck K, Uyttebroeck A, Vandenberghe P, Wlodarska I, Cloos J, Foà R, Speleman F, Cools J, Aerts S. Comprehensive analysis of transcriptome variation uncovers known and novel driver events in T-cell acute lymphoblastic leukemia. PLoS Genet 2013; 9:e1003997. [PMID: 24367274 PMCID: PMC3868543 DOI: 10.1371/journal.pgen.1003997] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/16/2013] [Indexed: 12/22/2022] Open
Abstract
RNA-seq is a promising technology to re-sequence protein coding genes for the identification of single nucleotide variants (SNV), while simultaneously obtaining information on structural variations and gene expression perturbations. We asked whether RNA-seq is suitable for the detection of driver mutations in T-cell acute lymphoblastic leukemia (T-ALL). These leukemias are caused by a combination of gene fusions, over-expression of transcription factors and cooperative point mutations in oncogenes and tumor suppressor genes. We analyzed 31 T-ALL patient samples and 18 T-ALL cell lines by high-coverage paired-end RNA-seq. First, we optimized the detection of SNVs in RNA-seq data by comparing the results with exome re-sequencing data. We identified known driver genes with recurrent protein altering variations, as well as several new candidates including H3F3A, PTK2B, and STAT5B. Next, we determined accurate gene expression levels from the RNA-seq data through normalizations and batch effect removal, and used these to classify patients into T-ALL subtypes. Finally, we detected gene fusions, of which several can explain the over-expression of key driver genes such as TLX1, PLAG1, LMO1, or NKX2-1; and others result in novel fusion transcripts encoding activated kinases (SSBP2-FER and TPM3-JAK2) or involving MLLT10. In conclusion, we present novel analysis pipelines for variant calling, variant filtering, and expression normalization on RNA-seq data, and successfully applied these for the detection of translocations, point mutations, INDELs, exon-skipping events, and expression perturbations in T-ALL. The quest for somatic mutations underlying oncogenic processes is a central theme in today's cancer research. High-throughput genomics approaches including amplicon re-sequencing, exome re-sequencing, full genome re-sequencing, and SNP arrays have contributed to cataloguing driver genes across cancer types. Thus far transcriptome sequencing by RNA-seq has been mainly used for the detection of fusion genes, while few studies have assessed its value for the combined detection of SNPs, INDELs, fusions, gene expression changes, and alternative transcript events. Here we apply RNA-seq to 49 T-ALL samples and perform a critical assessment of the bioinformatics pipelines and filters to identify each type of aberration. By comparing to exome re-sequencing, and by exploiting the catalogues of known cancer drivers, we identified many known and several novel driver genes in T-ALL. We also determined an optimal normalization strategy to obtain accurate gene expression levels and used these to identify over-expressed transcription factors that characterize different T-ALL subtypes. Finally, by PCR, cloning, and in vitro cellular assays we uncover new fusion genes that have consequences at the level of gene expression, oncogenic chimaeras, and tumor suppressor inactivation. In conclusion, we present the first RNA-seq data set across T-ALL patients and identify new driver events.
Collapse
Affiliation(s)
- Zeynep Kalender Atak
- Laboratory of Computational Biology, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Valentina Gianfelici
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, ‘Sapienza’ University of Rome, Rome, Italy
| | - Gert Hulselmans
- Laboratory of Computational Biology, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Kim De Keersmaecker
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
| | - Arun George Devasia
- Laboratory of Computational Biology, Center for Human Genetics, KU Leuven, Leuven, Belgium
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
| | - Ellen Geerdens
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
| | - Nicole Mentens
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
| | - Sabina Chiaretti
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, ‘Sapienza’ University of Rome, Rome, Italy
| | - Kaat Durinck
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Anne Uyttebroeck
- Pediatric Hemato-Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Peter Vandenberghe
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
| | - Iwona Wlodarska
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
| | - Jacqueline Cloos
- Pediatric Oncology/Hematology and Hematology, VU Medical Center, Amsterdam, The Netherlands
| | - Robin Foà
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, ‘Sapienza’ University of Rome, Rome, Italy
| | - Frank Speleman
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Jan Cools
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
- * E-mail: (JC); (SA)
| | - Stein Aerts
- Laboratory of Computational Biology, Center for Human Genetics, KU Leuven, Leuven, Belgium
- * E-mail: (JC); (SA)
| |
Collapse
|
216
|
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood malignancy and a leading case of childhood cancer death. The last decade has witnessed a transformation in our understanding of the genetic basis of ALL due to detailed integrative genomic profiling of large cohorts of childhood ALL. Initially using microarray based approaches, and more recently with next-generation sequencing, these studies have enabled more precise subclassification of ALL, and have shown that each ALL entity is characterized by constellations of structural and sequence mutations that typically perturb key cellular pathways including lymphoid development, cell cycle regulation, tumor suppression, Ras- and tyrosine kinase-driven signaling, and epigenetic regulation. Importantly, several of the newly identified genetic alterations have entered the clinic to improve diagnosis and risk stratification, and are being pursued as new targets for therapeutic intervention. Studies of ALL have also led the way in dissecting the subclonal heterogeneity of cancer, and have shown that individual patients commonly harbor multiple related but genetically distinct subclones, and that this genetically determined clonal heterogeneity is an important determinant of relapse. In addition, genome-wide profiling has identified inherited genetic variants that influence ALL risk. Ongoing studies are deploying detailed integrative genetic transcriptomic and epigenetic sequencing to comprehensively define the genomic landscape of ALL. This review describes the recent advances in our understanding of the genetics of ALL, with an emphasis on those alterations of key pathogenic or therapeutic importance.
Collapse
Affiliation(s)
- Charles G Mullighan
- Department of Pathology and the Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN.
| |
Collapse
|
217
|
Abstract
Our understanding of the pathogenesis of lymphoid malignancies has been transformed by next-generation sequencing. The studies in this review have used whole-genome, exome, and transcriptome sequencing to identify recurring structural genetic alterations and sequence mutations that target key cellular pathways in acute lymphoblastic leukemia (ALL) and the lymphomas. Although each tumor type is characterized by a unique genomic landscape, several cellular pathways are mutated in multiple tumor types-transcriptional regulation of differentiation, antigen receptor signaling, tyrosine kinase and Ras signaling, and epigenetic modifications-and individual genes are mutated in multiple tumors, notably TCF3, NOTCH1, MYD88, and BRAF. In addition to providing fundamental insights into tumorigenesis, these studies have also identified potential new markers for diagnosis, risk stratification, and therapeutic intervention. Several genetic alterations are intuitively "druggable" with existing agents, for example, kinase-activating lesions in high-risk B-cell ALL, NOTCH1 in both leukemia and lymphoma, and BRAF in hairy cell leukemia. Future sequencing efforts are required to comprehensively define the genetic basis of all lymphoid malignancies, examine the relative roles of germline and somatic variation, dissect the genetic basis of clonal heterogeneity, and chart a course for clinical sequencing and translation to improved therapeutic outcomes.
Collapse
|
218
|
Independent prognostic value of BCR-ABL1-like signature and IKZF1 deletion, but not high CRLF2 expression, in children with B-cell precursor ALL. Blood 2013; 122:2622-9. [PMID: 23974192 DOI: 10.1182/blood-2012-10-462358] [Citation(s) in RCA: 211] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Most relapses in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL) are not predicted using current prognostic features. Here, we determined the co-occurrence and independent prognostic relevance of 3 recently identified prognostic features: BCR-ABL1-like gene signature, deletions in IKZF1, and high CRLF2 messenger RNA expression (CRLF2-high). These features were determined in 4 trials representing 1128 children with ALL: DCOG ALL-8, ALL9, ALL10, and Cooperative ALL (COALL)-97/03. BCR-ABL1-like, IKZF1-deleted, and CRLF2-high cases constitute 33.7% of BCR-ABL1-negative, MLL wild-type BCP-ALL cases, of which BCR-ABL1-like and IKZF1 deletion (co)occurred most frequently. Higher cumulative incidence of relapse was found for BCR-ABL1-like and IKZF1-deleted, but not CRLF2-high, cases relative to remaining BCP-ALL cases, reflecting the observations in each of the cohorts analyzed separately. No relapses occurred among cases with CRLF2-high as single feature, whereas 62.9% of all relapses in BCR-ABL1-negative, MLL wild-type BCP-ALL occurred in cases with BCR-ABL1-like signature and/or IKZF1 deletion. Both the BCR-ABL1-like signature and IKZF1 deletions were prognostic features independent of conventional prognostic markers in a multivariate model, and both remained prognostic among cases with intermediate minimal residual disease. The BCR-ABL1-like signature and an IKZF1 deletion, but not CRLF2-high, are prognostic factors and are clinically of importance to identify high-risk patients who require more intensive and/or alternative therapies.
Collapse
|
219
|
Cario G, Rhein P, Mitlöhner R, Zimmermann M, Bandapalli OR, Romey R, Moericke A, Ludwig WD, Ratei R, Muckenthaler MU, Kulozik AE, Schrappe M, Stanulla M, Karawajew L. High CD45 surface expression determines relapse risk in children with precursor B-cell and T-cell acute lymphoblastic leukemia treated according to the ALL-BFM 2000 protocol. Haematologica 2013; 99:103-10. [PMID: 23911702 DOI: 10.3324/haematol.2013.090225] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Further improvement of outcome in childhood acute lymphoblastic leukemia could be achieved by identifying additional high-risk patients who may benefit from intensified treatment. We earlier identified PTPRC (CD45) gene expression as a potential new stratification marker and now analyzed the prognostic relevance of CD45 protein expression. CD45 was measured by flow cytometry in 1065 patients treated according to the ALL-BFM-2000 protocol. The 75(th) percentile was used as cut-off to distinguish a CD45-high from a CD45-low group. As mean CD45 expression was significantly higher in T-cell acute lymphoblastic leukemia than in B-cell-precursor acute lymphoblastic leukemia (P<0.0001), the analysis was performed separately in both groups. In B-cell-precursor acute lymphoblastic leukemia we observed a significant association of a high CD45 expression with older age, high initial white blood cell count, ETV6/RUNX1 negativity, absence of high hyperdiploidy (P<0.0001), MLL/AF4 positivity (P=0.002), BCR/ABL1 positivity (P=0.007), prednisone poor response (P=0.002) and minimal residual disease (P<0.0001). In T-cell acute lymphoblastic leukemia we observed a significant association with initial white blood cell count (P=0.0003), prednisone poor response (P=0.01), and minimal residual disease (P=0.02). Compared to CD45-low patients, CD45-high patients had a lower event-free survival rate (B-cell-precursor acute lymphoblastic leukemia: 72 ± 3% versus 86 ± 1%, P<0.0001; T-cell acute lymphoblastic leukemia: 60 ± 8% versus 78 ± 4%, P=0.02), which was mainly attributable to a higher cumulative relapse incidence (B-cell-precursor acute lymphoblastic leukemia: 22 ± 3% versus 11 ± 1%, P<0.0001; T-cell acute lymphoblastic leukemia: 31 ± 8% versus 11 ± 3%, P=0.003) and kept its significance in multivariate analysis considering sex, age, initial white blood cell count, and minimal residual disease in B-cell-precursor- and T-cell acute lymphoblastic leukemia, and additionally presence of ETV6/RUNX1, MLL/AF4 and BCR/ABL1 rearrangements in B-cell-precursor acute lymphoblastic leukemia (P=0.002 and P=0.025, respectively). Consideration of CD45 expression may serve as an additional stratification tool in BFM-based protocols. (ClinicalTrials.gov identifier: NCT00430118).
Collapse
|
220
|
Abstract
The SH2B adaptor protein 3 (SH2B3) gene encodes a negative regulator of cytokine signaling with a critical role in the homeostasis of hematopoietic stem cells and lymphoid progenitors. Here, we report the identification of germline homozygous SH2B3 mutations in 2 siblings affected with developmental delay and autoimmunity, one in whom B-precursor acute lymphoblastic leukemia (ALL) developed. Mechanistically, loss of SH2B3 increases Janus kinase-signal transducer and activator of transcription signaling, promotes lymphoid cell proliferation, and accelerates leukemia development in a mouse model of NOTCH1-induced ALL. Moreover, extended mutation analysis showed homozygous somatic mutations in SH2B3 in 2 of 167 ALLs analyzed. Overall, these results demonstrate a Knudson tumor suppressor role for SH2B3 in the pathogenesis of ALL and highlight a possible link between genetic predisposition factors in the pathogenesis of autoimmunity and leukemogenesis.
Collapse
|
221
|
Menet CJ, Rompaey LV, Geney R. Advances in the discovery of selective JAK inhibitors. PROGRESS IN MEDICINAL CHEMISTRY 2013; 52:153-223. [PMID: 23384668 DOI: 10.1016/b978-0-444-62652-3.00004-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
In this review, we describe the current knowledge of the biology of the JAKs. The JAK family comprises the four nonreceptor tyrosine kinases JAK1, JAK2, JAK3, and Tyk2, all key players in the signal transduction from cytokine receptors to transcription factor activation. We also review the progresses made towards the optimization of JAK inhibitors and the importance of their selectivity profile. Indeed, the full array of many medicinal chemistry enabling tools (HTS, X-ray crystallography, scaffold morphing, etc.) has been deployed to successfully design molecules that discriminate among JAK family and other kinases. While the first JAK inhibitor was launched in 2011, this review also summarizes the status of several other small-molecule JAK inhibitors currently in development to treat arthritis, psoriasis, organ rejection, and multiple cancer types.
Collapse
|
222
|
Ribeiro D, Melão A, Barata JT. IL-7R-mediated signaling in T-cell acute lymphoblastic leukemia. Adv Biol Regul 2013; 53:211-222. [PMID: 23234870 DOI: 10.1016/j.jbior.2012.10.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 10/09/2012] [Indexed: 06/01/2023]
Abstract
Interleukin-7 (IL-7), a cytokine produced in the bone marrow, thymus and other organs, is mandatory for normal human T-cell development and peripheral homeostasis. Different studies, including phase I clinical trials, have indicated the potential therapeutic value of recombinant IL-7 in the context of anti-cancer immunotherapy and as a booster of immune reconstitution. However, the two main pathways activated by IL-7, JAK/STAT5 and PI3K/Akt/mTOR, have both been implicated in cancer and there is considerable evidence that IL-7 and its receptor (IL-7R), formed by IL-7Rα (encoded by IL7R) and γc, may partake in T-cell acute lymphoblastic leukemia (T-ALL) development. In this context, the most compelling data comes from recent studies demonstrating that around 10% of T-ALL patients display IL7R gain-of-function mutations leading, in most cases, to disulfide bond-dependent homodimerization of two mutant receptors and consequent constitutive activation of downstream signaling, with ensuing cell transformation in vitro and tumorigenic ability in vivo. Here, we review the data on the involvement of IL-7 and IL-7R in T-ALL, further discussing the peculiarities of IL-7R-mediated signaling in human leukemia T-cells that may be of therapeutic value, namely regarding the potential use of PI3K and mTOR pharmacological inhibitors.
Collapse
Affiliation(s)
- Daniel Ribeiro
- Instituto de Medicina Molecular, Faculdade de Medicina, Unversidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | | | | |
Collapse
|
223
|
|
224
|
Walsh STR. Structural insights into the common γ-chain family of cytokines and receptors from the interleukin-7 pathway. Immunol Rev 2013; 250:303-16. [PMID: 23046137 DOI: 10.1111/j.1600-065x.2012.01160.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Over the past 13 years, numerous crystal structures of complexes of the common γ-chain (γ(c)) cytokine receptors and their cytokines have been solved. Even with the remarkable progress in the structural biology of γ(c) receptors and their cytokines or interleukins, there are valuable lessons to be learned from the structural and biophysical studies of interleukin-7 (IL-7) and its α-receptor (IL-7Rα) and comparisons with other γ(c) family members. The structure of the IL-7/IL-7Rα complex teaches that interfaces between the γ(c) interleukins and their receptors can vary in size, polarity, and specificity, and that significant conformational changes might be necessary for complexes of interleukins and their receptors to bind the shared, activating γ(c) receptor. Binding, kinetic, and thermodynamic studies of IL-7 and IL-7Rα show that glycosylation and electrostatics can be important to interactions between interleukins and their receptor, even where the glycans and charged residues are distant from the interface. The structure of the IL-7Rα homodimer is a reminder that often-ignored non-activating complexes likely perform roles just as important to signaling as activating complexes. And last but not least, the structural and biophysical studies help explain and potentially treat the diseases caused by aberrant IL-7 signaling.
Collapse
Affiliation(s)
- Scott T R Walsh
- Department of Cell Biology and Molecular Genetics, Institute for Bioscience and Biotechnology Research, W. M. Keck Laboratory for Structural Biology, Rockville, MD, USA.
| |
Collapse
|
225
|
Huh HJ, Lee SH, Yoo KH, Sung KW, Koo HH, Jang JH, Kim K, Kim SJ, Kim WS, Jung CW, Lee KO, Kim SH, Kim HJ. Gene mutation profiles and prognostic implications in Korean patients with T-lymphoblastic leukemia. Ann Hematol 2013; 92:635-44. [PMID: 23354995 DOI: 10.1007/s00277-012-1664-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 12/17/2012] [Indexed: 11/29/2022]
Abstract
Genetic alterations implicated in the leukemogenesis of T cell acute lymphoblastic leukemia (T-ALL) have been identified in recent years. In this study, we investigated gene mutation profiles and prognostic implications in a series of Korean T-ALL patients. The study patients were 29 Korean patients with T-ALL; 13 adults (45 %) and 16 children (55 %; male-to-female ratio, 25:4). Clinical, hematologic, and cytogenetic findings were reviewed. We performed mutation analyses for NOTCH1, FBXW7, PHF6, and IL7R genes and survival analyses according to the mutational status. Gene mutations were identified in 66 % of the patients in our series (19/29). Eighteen patients (62 %) had NOTCH1/FBXW7 mutations. Sixteen patients (55 %) had NOTCH1 mutations including nine novel mutations, and eight patients (28 %) had known FBXW7 mutations. Eight patients (28 %; six males and two females) had PHF6 mutations including four novel mutations. Three patients (10 %) had IL7R mutations, which were all novel in-frame insertion or deletion-insertions. The gene mutation profile combined with cytogenetics and FISH study for the p16 gene detected genetic aberrations in 90 % of patients (26/29). There was no significant difference in the frequency of gene mutations between the pediatric and adult patients with T-ALL. Survival analyses suggested a favorable prognostic implication of NOTCH1 mutations in adult T-ALL. Gene mutation studies for NOTCH1, FBXW7, PHF6, and IL7R could detect genetic alterations in a majority of Korean T-ALL patients with novel mutations. We observed similar mutation profiles between adult and pediatric T-ALL, and a favorable prognostic implication of NOTCH1 mutations in adult T-ALL.
Collapse
Affiliation(s)
- Hee Jae Huh
- Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Bandapalli OR, Zimmermann M, Kox C, Stanulla M, Schrappe M, Ludwig WD, Koehler R, Muckenthaler MU, Kulozik AE. NOTCH1 activation clinically antagonizes the unfavorable effect of PTEN inactivation in BFM-treated children with precursor T-cell acute lymphoblastic leukemia. Haematologica 2013; 98:928-36. [PMID: 23349303 DOI: 10.3324/haematol.2012.073585] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Despite improvements in treatment results for pediatric T-cell acute lymphoblastic leukemia, approximately 20% of patients relapse with dismal prognosis. PTEN inactivation and NOTCH1 activation are known frequent leukemogenic events but their effect on outcome is still controversial. We analyzed the effect of PTEN inactivation and its interaction with NOTCH1 activation on treatment response and long-term outcome in 301 ALL-BFM treated children with T-cell acute lymphoblastic leukemia. We identified PTEN mutations in 52 of 301 (17.3%) of patients. In univariate analyses this was significantly associated with increased resistance to induction chemotherapy and a trend towards poor long-term outcome. By contrast, patients with inactivating PTEN and activating NOTCH1 mutations showed marked sensitivity to induction treatment and excellent long-term outcome, which was similar to patients with NOTCH1 mutations only, and more favorable than in patients with PTEN mutations only. Notably, in the subgroup of patients with a prednisone- and minimal residual disease (MRD)-response based medium risk profile, PTEN-mutations without co-existing NOTCH1-mutations represented an MRD-independent highly significant high-risk biomarker. Mutations of PTEN highly significantly indicate a poor prognosis in T-ALL patients who have been stratified to the medium risk group of the BFM-protocol. This effect is clinically neutralized by NOTCH1 mutations. Although these results have not yet been explained by an obvious molecular mechanism, they contribute to the development of new molecularly defined stratification algorithms. Furthermore, these data have unexpected potential implications for the development of NOTCH1 inhibitors in the treatment of T-cell acute lymphoblastic leukemia in general, and in those with a combination of PTEN and NOTCH1 mutations in particular.
Collapse
Affiliation(s)
- Obul R Bandapalli
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Affiliation(s)
- John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892-1616, USA.
| | | | | |
Collapse
|
228
|
Gatt ME, Ben-Yehuda D, Izraeli S. Lymphoid leukemias. Clin Immunol 2013. [DOI: 10.1016/b978-0-7234-3691-1.00092-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
229
|
Loh ML, Mullighan CG. Advances in the genetics of high-risk childhood B-progenitor acute lymphoblastic leukemia and juvenile myelomonocytic leukemia: implications for therapy. Clin Cancer Res 2012; 18:2754-67. [PMID: 22589484 DOI: 10.1158/1078-0432.ccr-11-1936] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hematologic malignancies of childhood comprise the most common childhood cancers. These neoplasms derive from the pathologic clonal expansion of an abnormal cancer-initiating cell and span a diverse spectrum of phenotypes, including acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), myeloproliferative neoplasms (MPN), and myelodysplastic syndromes (MDS). Expansion of immature lymphoid or myeloid blasts with suppression of normal hematopoiesis is the hallmark of ALL and AML, whereas MPN is associated with proliferation of 1 or more lineages that retain the ability to differentiate, and MDS is characterized by abnormal hematopoiesis and cytopenias. The outcomes for children with the most common childhood cancer, B-progenitor ALL (B-ALL), in general, is quite favorable, in contrast to children affected by myeloid malignancies. The advent of highly sensitive genomic technologies reveals the remarkable genetic complexity of multiple subsets of high-risk B-progenitor ALL, in contrast to a somewhat simpler model of myeloid neoplasms, although a number of recently discovered alterations displayed by both types of malignancies may lead to common therapeutic approaches. This review outlines recent advances in our understanding of the genetic underpinnings of high-risk B-ALL and juvenile myelomonocytic leukemia, an overlap MPN/MDS found exclusively in children, and we also discuss novel therapeutic approaches that are currently being tested in clinical trials. Recent insights into the clonal heterogeneity of leukemic samples and the implications for diagnostic and therapeutic approaches are also discussed.
Collapse
Affiliation(s)
- Mignon L Loh
- Department of Pediatrics and the Helen Diller Comprehensive Cancer Center, Benioff Children's Hospital, University of California, San Francisco, San Francisco, California, USA
| | | |
Collapse
|
230
|
Maude SL, Tasian SK, Vincent T, Hall JW, Sheen C, Roberts KG, Seif AE, Barrett DM, Chen IM, Collins JR, Mullighan CG, Hunger SP, Harvey RC, Willman CL, Fridman JS, Loh ML, Grupp SA, Teachey DT. Targeting JAK1/2 and mTOR in murine xenograft models of Ph-like acute lymphoblastic leukemia. Blood 2012; 120:3510-8. [PMID: 22955920 PMCID: PMC3482861 DOI: 10.1182/blood-2012-03-415448] [Citation(s) in RCA: 209] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 08/21/2012] [Indexed: 12/24/2022] Open
Abstract
CRLF2 rearrangements, JAK1/2 point mutations, and JAK2 fusion genes have been identified in Philadelphia chromosome (Ph)-like acute lymphoblastic leukemia (ALL), a recently described subtype of pediatric high-risk B-precursor ALL (B-ALL) which exhibits a gene expression profile similar to Ph-positive ALL and has a poor prognosis. Hyperactive JAK/STAT and PI3K/mammalian target of rapamycin (mTOR) signaling is common in this high-risk subset. We, therefore, investigated the efficacy of the JAK inhibitor ruxolitinib and the mTOR inhibitor rapamycin in xenograft models of 8 pediatric B-ALL cases with and without CRLF2 and JAK genomic lesions. Ruxolitinib treatment yielded significantly lower peripheral blast counts compared with vehicle (P < .05) in 6 of 8 human leukemia xenografts and lower splenic blast counts (P < .05) in 8 of 8 samples. Enhanced responses to ruxolitinib were observed in samples harboring JAK-activating lesions and higher levels of STAT5 phosphorylation. Rapamycin controlled leukemia burden in all 8 B-ALL samples. Survival analysis of 2 representative B-ALL xenografts demonstrated prolonged survival with rapamycin treatment compared with vehicle (P < .01). These data demonstrate preclinical in vivo efficacy of ruxolitinib and rapamycin in this high-risk B-ALL subtype, for which novel treatments are urgently needed, and highlight the therapeutic potential of targeted kinase inhibition in Ph-like ALL.
Collapse
Affiliation(s)
- Shannon L Maude
- Division of Oncology, The Children's Hospital of Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Small sizes and indolent evolutionary dynamics challenge the potential role of P2RY8-CRLF2-harboring clones as main relapse-driving force in childhood ALL. Blood 2012; 120:5134-42. [PMID: 23091296 DOI: 10.1182/blood-2012-07-443218] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The P2RY8-CRLF2 fusion defines a particular relapse-prone subset of childhood acute lymphoblastic leukemia (ALL) in Italian Association of Pediatric Hematology and Oncology Berlin-Frankfurt-Münster (AIEOP-BFM) 2000 protocols. To investigate whether and to what extent different clone sizes influence disease and relapse development, we quantified the genomic P2RY8-CRLF2 fusion product and correlated it with the corresponding CRLF2 expression levels in patients enrolled in the BFM-ALL 2000 protocol in Austria. Of 268 cases without recurrent chromosomal translocations and high hyperdiploidy, representing approximately 50% of all cases, 67 (25%) were P2RY8-CRLF2 positive. The respective clone sizes were ≥ 20% in 27% and < 20% in 73% of them. The cumulative incidence of relapse of the entire fusion-positive group was clone size independent and significantly higher than that of the fusion-negative group (35% ± 8% vs 13% ± 3%, P = .008) and primarily confined to the non-high-risk group. Of 22 P2RY8-CRLF2-positive diagnosis/relapse pairs, only 4/8 had the fusion-positive dominant clone conserved at relapse, whereas none of the original 14 fusion-positive small clones reappeared as the dominant relapse clone. We conclude that the majority of P2RY8-CRLF2-positive clones are small at diagnosis and virtually never generate a dominant relapse clone. Our findings therefore suggest that P2RY8-CRLF2-positive clones do not have the necessary proliferative or selective advantage to evolve into a disease-relevant relapse clone.
Collapse
|
232
|
Demehri S, Turkoz A, Manivasagam S, Yockey LJ, Turkoz M, Kopan R. Elevated epidermal thymic stromal lymphopoietin levels establish an antitumor environment in the skin. Cancer Cell 2012; 22:494-505. [PMID: 23079659 PMCID: PMC3480666 DOI: 10.1016/j.ccr.2012.08.017] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 06/13/2012] [Accepted: 08/21/2012] [Indexed: 12/31/2022]
Abstract
Thymic Stromal Lymphopoietin (TSLP), a cytokine implicated in induction of T helper 2 (Th2)-mediated allergic inflammation, has recently been shown to stimulate solid tumor growth and metastasis. Conversely, studying mice with clonal loss of Notch signaling in their skin revealed that high levels of TSLP released by barrier-defective skin caused a severe inflammation, resulting in gradual elimination of Notch-deficient epidermal clones and resistance to skin tumorigenesis. We found CD4(+) T cells to be both required and sufficient to mediate these effects of TSLP. Importantly, TSLP overexpression in wild-type skin also caused resistance to tumorigenesis, confirming that TSLP functions as a tumor suppressor in the skin.
Collapse
Affiliation(s)
- Shadmehr Demehri
- Department of Developmental Biology and Division of Dermatology, Washington University School of Medicine, Box 8103, 660 South Euclid Avenue, Saint Louis, Missouri 63110-1095, USA
- Department of Internal Medicine, St. Luke’s Hospital, 232 South Woods Mill Road, Chesterfield, Missouri 63017, USA
- Authors for correspondence: ,
| | - Ahu Turkoz
- Department of Developmental Biology and Division of Dermatology, Washington University School of Medicine, Box 8103, 660 South Euclid Avenue, Saint Louis, Missouri 63110-1095, USA
| | - Sindhu Manivasagam
- Department of Developmental Biology and Division of Dermatology, Washington University School of Medicine, Box 8103, 660 South Euclid Avenue, Saint Louis, Missouri 63110-1095, USA
| | - Laura J. Yockey
- Department of Developmental Biology and Division of Dermatology, Washington University School of Medicine, Box 8103, 660 South Euclid Avenue, Saint Louis, Missouri 63110-1095, USA
| | - Mustafa Turkoz
- Department of Developmental Biology and Division of Dermatology, Washington University School of Medicine, Box 8103, 660 South Euclid Avenue, Saint Louis, Missouri 63110-1095, USA
| | - Raphael Kopan
- Department of Developmental Biology and Division of Dermatology, Washington University School of Medicine, Box 8103, 660 South Euclid Avenue, Saint Louis, Missouri 63110-1095, USA
- Authors for correspondence: ,
| |
Collapse
|
233
|
Liang WS, Craig DW, Carpten J, Borad MJ, Demeure MJ, Weiss GJ, Izatt T, Sinari S, Christoforides A, Aldrich J, Kurdoglu A, Barrett M, Phillips L, Benson H, Tembe W, Braggio E, Kiefer JA, Legendre C, Posner R, Hostetter GH, Baker A, Egan JB, Han H, Lake D, Stites EC, Ramanathan RK, Fonseca R, Stewart AK, Von Hoff D. Genome-wide characterization of pancreatic adenocarcinoma patients using next generation sequencing. PLoS One 2012; 7:e43192. [PMID: 23071490 PMCID: PMC3468610 DOI: 10.1371/journal.pone.0043192] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 07/19/2012] [Indexed: 12/24/2022] Open
Abstract
Pancreatic adenocarcinoma (PAC) is among the most lethal malignancies. While research has implicated multiple genes in disease pathogenesis, identification of therapeutic leads has been difficult and the majority of currently available therapies provide only marginal benefit. To address this issue, our goal was to genomically characterize individual PAC patients to understand the range of aberrations that are occurring in each tumor. Because our understanding of PAC tumorigenesis is limited, evaluation of separate cases may reveal aberrations, that are less common but may provide relevant information on the disease, or that may represent viable therapeutic targets for the patient. We used next generation sequencing to assess global somatic events across 3 PAC patients to characterize each patient and to identify potential targets. This study is the first to report whole genome sequencing (WGS) findings in paired tumor/normal samples collected from 3 separate PAC patients. We generated on average 132 billion mappable bases across all patients using WGS, and identified 142 somatic coding events including point mutations, insertion/deletions, and chromosomal copy number variants. We did not identify any significant somatic translocation events. We also performed RNA sequencing on 2 of these patients' tumors for which tumor RNA was available to evaluate expression changes that may be associated with somatic events, and generated over 100 million mapped reads for each patient. We further performed pathway analysis of all sequencing data to identify processes that may be the most heavily impacted from somatic and expression alterations. As expected, the KRAS signaling pathway was the most heavily impacted pathway (P<0.05), along with tumor-stroma interactions and tumor suppressive pathways. While sequencing of more patients is needed, the high resolution genomic and transcriptomic information we have acquired here provides valuable information on the molecular composition of PAC and helps to establish a foundation for improved therapeutic selection.
Collapse
Affiliation(s)
- Winnie S. Liang
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - David W. Craig
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - John Carpten
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | | | - Michael J. Demeure
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Virginia G. Piper Cancer Center Clinical Trials, Scottsdale Healthcare, Scottsdale, Arizona, United States of America
| | - Glen J. Weiss
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Virginia G. Piper Cancer Center Clinical Trials, Scottsdale Healthcare, Scottsdale, Arizona, United States of America
| | - Tyler Izatt
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Shripad Sinari
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Alexis Christoforides
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Jessica Aldrich
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Ahmet Kurdoglu
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Michael Barrett
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Lori Phillips
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Hollie Benson
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Waibhav Tembe
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | | | - Jeffrey A. Kiefer
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Christophe Legendre
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Richard Posner
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Galen H. Hostetter
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Angela Baker
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Jan B. Egan
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Haiyong Han
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Douglas Lake
- Arizona State University, Tempe, Arizona, United States of America
| | - Edward C. Stites
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Ramesh K. Ramanathan
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Virginia G. Piper Cancer Center Clinical Trials, Scottsdale Healthcare, Scottsdale, Arizona, United States of America
| | - Rafael Fonseca
- Mayo Clinic, Scottsdale, Arizona, United States of America
| | | | - Daniel Von Hoff
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Mayo Clinic, Scottsdale, Arizona, United States of America
- Virginia G. Piper Cancer Center Clinical Trials, Scottsdale Healthcare, Scottsdale, Arizona, United States of America
- * E-mail:
| |
Collapse
|
234
|
Liang WS, Craig DW, Carpten J, Borad MJ, Demeure MJ, Weiss GJ, Izatt T, Sinari S, Christoforides A, Aldrich J, Kurdoglu A, Barrett M, Phillips L, Benson H, Tembe W, Braggio E, Kiefer JA, Legendre C, Posner R, Hostetter GH, Baker A, Egan JB, Han H, Lake D, Stites EC, Ramanathan RK, Fonseca R, Stewart AK, Von Hoff D. Genome-wide characterization of pancreatic adenocarcinoma patients using next generation sequencing. PLoS One 2012. [PMID: 23071490 DOI: 10.137/journal.pone.0043192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pancreatic adenocarcinoma (PAC) is among the most lethal malignancies. While research has implicated multiple genes in disease pathogenesis, identification of therapeutic leads has been difficult and the majority of currently available therapies provide only marginal benefit. To address this issue, our goal was to genomically characterize individual PAC patients to understand the range of aberrations that are occurring in each tumor. Because our understanding of PAC tumorigenesis is limited, evaluation of separate cases may reveal aberrations, that are less common but may provide relevant information on the disease, or that may represent viable therapeutic targets for the patient. We used next generation sequencing to assess global somatic events across 3 PAC patients to characterize each patient and to identify potential targets. This study is the first to report whole genome sequencing (WGS) findings in paired tumor/normal samples collected from 3 separate PAC patients. We generated on average 132 billion mappable bases across all patients using WGS, and identified 142 somatic coding events including point mutations, insertion/deletions, and chromosomal copy number variants. We did not identify any significant somatic translocation events. We also performed RNA sequencing on 2 of these patients' tumors for which tumor RNA was available to evaluate expression changes that may be associated with somatic events, and generated over 100 million mapped reads for each patient. We further performed pathway analysis of all sequencing data to identify processes that may be the most heavily impacted from somatic and expression alterations. As expected, the KRAS signaling pathway was the most heavily impacted pathway (P<0.05), along with tumor-stroma interactions and tumor suppressive pathways. While sequencing of more patients is needed, the high resolution genomic and transcriptomic information we have acquired here provides valuable information on the molecular composition of PAC and helps to establish a foundation for improved therapeutic selection.
Collapse
Affiliation(s)
- Winnie S Liang
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Martelli AM, Chiarini F, Evangelisti C, Cappellini A, Buontempo F, Bressanin D, Fini M, McCubrey JA. Two hits are better than one: targeting both phosphatidylinositol 3-kinase and mammalian target of rapamycin as a therapeutic strategy for acute leukemia treatment. Oncotarget 2012; 3:371-94. [PMID: 22564882 PMCID: PMC3380573 DOI: 10.18632/oncotarget.477] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR) are two key components of the PI3K/Akt/mTOR signaling pathway. This signal transduction cascade regulates a wide range of physiological cell processes, that include differentiation, proliferation, apoptosis, autophagy, metabolism, motility, and exocytosis. However, constitutively active PI3K/Akt/mTOR signaling characterizes many types of tumors where it negatively influences response to therapeutic treatments. Hence, targeting PI3K/Akt/mTOR signaling with small molecule inhibitors may improve cancer patient outcome. The PI3K/Akt/mTOR signaling cascade is overactive in acute leukemias, where it correlates with enhanced drug-resistance and poor prognosis. The catalytic sites of PI3K and mTOR share a high degree of sequence homology. This feature has allowed the synthesis of ATP-competitive compounds targeting the catalytic site of both kinases. In preclinical models, dual PI3K/mTOR inhibitors displayed a much stronger cytotoxicity against acute leukemia cells than either PI3K inhibitors or allosteric mTOR inhibitors, such as rapamycin. At variance with rapamycin, dual PI3K/mTOR inhibitors targeted both mTOR complex 1 and mTOR complex 2, and inhibited the rapamycin-resistant phosphorylation of eukaryotic initiation factor 4E-binding protein 1, resulting in a marked inhibition of oncogenic protein translation. Therefore, they strongly reduced cell proliferation and induced an important apoptotic response. Here, we reviewed the evidence documenting that dual PI3K/mTOR inhibitors may represent a promising option for future targeted therapies of acute leukemia patients.
Collapse
Affiliation(s)
- Alberto M Martelli
- Department of Human Anatomy, University of Bologna, Cellular Signalling Laboratory, Bologna, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
236
|
Perturbation of fetal liver hematopoietic stem and progenitor cell development by trisomy 21. Proc Natl Acad Sci U S A 2012; 109:17579-84. [PMID: 23045701 DOI: 10.1073/pnas.1211405109] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The 40-fold increase in childhood megakaryocyte-erythroid and B-cell leukemia in Down syndrome implicates trisomy 21 (T21) in perturbing fetal hematopoiesis. Here, we show that compared with primary disomic controls, primary T21 fetal liver (FL) hematopoietic stem cells (HSC) and megakaryocyte-erythroid progenitors are markedly increased, whereas granulocyte-macrophage progenitors are reduced. Commensurately, HSC and megakaryocyte-erythroid progenitors show higher clonogenicity, with increased megakaryocyte, megakaryocyte-erythroid, and replatable blast colonies. Biased megakaryocyte-erythroid-primed gene expression was detected as early as the HSC compartment. In lymphopoiesis, T21 FL lymphoid-primed multipotential progenitors and early lymphoid progenitor numbers are maintained, but there was a 10-fold reduction in committed PreproB-lymphoid progenitors and the functional B-cell potential of HSC and early lymphoid progenitor is severely impaired, in tandem with reduced early lymphoid gene expression. The same pattern was seen in all T21 FL samples and no samples had GATA1 mutations. Therefore, T21 itself causes multiple distinct defects in FL myelo- and lymphopoiesis.
Collapse
|
237
|
Van Vlierberghe P, Ferrando A. The molecular basis of T cell acute lymphoblastic leukemia. J Clin Invest 2012; 122:3398-406. [PMID: 23023710 DOI: 10.1172/jci61269] [Citation(s) in RCA: 375] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
T cell acute lymphoblastic leukemias (T-ALLs) arise from the malignant transformation of hematopoietic progenitors primed toward T cell development, as result of a multistep oncogenic process involving constitutive activation of NOTCH signaling and genetic alterations in transcription factors, signaling oncogenes, and tumor suppressors. Notably, these genetic alterations define distinct molecular groups of T-ALL with specific gene expression signatures and clinicobiological features. This review summarizes recent advances in our understanding of the molecular genetics of T-ALL.
Collapse
Affiliation(s)
- Pieter Van Vlierberghe
- Institute for Cancer Genetics, Department of Pathology, Columbia University Medical Center, New York, New York 10032, USA
| | | |
Collapse
|
238
|
Mullighan CG. Molecular genetics of B-precursor acute lymphoblastic leukemia. J Clin Invest 2012; 122:3407-15. [PMID: 23023711 DOI: 10.1172/jci61203] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
B-precursor acute lymphoblastic leukemia (B-ALL) is the most common childhood tumor and the leading cause of cancer-related death in children and young adults. The majority of B-ALL cases are aneuploid or harbor recurring structural chromosomal rearrangements that are important initiating events in leukemogenesis but are insufficient to explain the biology and heterogeneity of disease. Recent studies have used microarrays and sequencing to comprehensively identify all somatic genetic alterations in acute lymphoblastic leukemia (ALL). These studies have identified cryptic or submicroscopic genetic alterations that define new ALL subtypes, cooperate with known chromosomal rearrangements, and influence prognosis. This article reviews these advances, discusses results from ongoing second-generation sequencing studies of ALL, and highlights challenges and opportunities for future genetic profiling approaches.
Collapse
Affiliation(s)
- Charles G Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, USA.
| |
Collapse
|
239
|
Barrett D, Brown VI, Grupp SA, Teachey DT. Targeting the PI3K/AKT/mTOR signaling axis in children with hematologic malignancies. Paediatr Drugs 2012; 14:299-316. [PMID: 22845486 PMCID: PMC4214862 DOI: 10.2165/11594740-000000000-00000] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The phosphatidylinositiol 3-kinase (PI3K), AKT, mammalian target of rapamycin (mTOR) signaling pathway (PI3K/AKT/mTOR) is frequently dysregulated in disorders of cell growth and survival, including a number of pediatric hematologic malignancies. The pathway can be abnormally activated in childhood acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), and chronic myelogenous leukemia (CML), as well as in some pediatric lymphomas and lymphoproliferative disorders. Most commonly, this abnormal activation occurs as a consequence of constitutive activation of AKT, providing a compelling rationale to target this pathway in many of these conditions. A variety of agents, beginning with the rapamycin analogue (rapalog) sirolimus, have been used successfully to target this pathway in a number of pediatric hematologic malignancies. Rapalogs demonstrate significant preclinical activity against ALL, which has led to a number of clinical trials. Moreover, rapalogs can synergize with a number of conventional cytotoxic agents and overcome pathways of chemotherapeutic resistance for drugs commonly used in ALL treatment, including methotrexate and corticosteroids. Based on preclinical data, rapalogs are also being studied in AML, CML, and non-Hodgkin's lymphoma. Recently, significant progress has been made using rapalogs to treat pre-malignant lymphoproliferative disorders, including the autoimmune lymphoproliferative syndrome (ALPS); complete remissions in children with otherwise therapy-resistant disease have been seen. Rapalogs only block one component of the pathway (mTORC1), and newer agents are under preclinical and clinical development that can target different and often multiple protein kinases in the PI3K/AKT/mTOR pathway. Most of these agents have been tolerated in early-phase clinical trials. A number of PI3K inhibitors are under investigation. Of note, most of these also target other protein kinases. Newer agents are under development that target both mTORC1 and mTORC2, mTORC1 and PI3K, and the triad of PI3K, mTORC1, and mTORC2. Preclinical data suggest these dual- and multi-kinase inhibitors are more potent than rapalogs against many of the aforementioned hematologic malignancies. Two classes of AKT inhibitors are under development, the alkyl-lysophospholipids (APLs) and small molecule AKT inhibitors. Both classes have agents currently in clinical trials. A number of drugs are in development that target other components of the pathway, including eukaryotic translation initiation factor (eIF) 4E (eIF4E) and phosphoinositide-dependent protein kinase 1 (PDK1). Finally, a number of other key signaling pathways interact with PI3K/AKT/mTOR, including Notch, MNK, Syk, MAPK, and aurora kinase. These alternative pathways are being targeted alone and in combination with PI3K/AKT/mTOR inhibitors with promising preclinical results in pediatric hematologic malignancies. This review provides a comprehensive overview of the abnormalities in the PI3K/AKT/mTOR signaling pathway in pediatric hematologic malignancies, the agents that are used to target this pathway, and the results of preclinical and clinical trials, using those agents in childhood hematologic cancers.
Collapse
Affiliation(s)
- David Barrett
- Department of Pediatrics, Division of Oncology, Children’s
Hospital of Philadelphia, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
| | - Valerie I. Brown
- Department of Pediatrics, Division of Oncology, Children’s
Hospital of Philadelphia, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
| | - Stephan A. Grupp
- Department of Pediatrics, Division of Oncology, Children’s
Hospital of Philadelphia, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
| | - David T. Teachey
- Department of Pediatrics, Division of Oncology, Children’s
Hospital of Philadelphia, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
- Department of Pediatrics, Division of Hematology, Children’s
Hospital of Philadelphia, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
| |
Collapse
|
240
|
JAK2 the future: therapeutic strategies for JAK-dependent malignancies. Trends Pharmacol Sci 2012; 33:574-82. [PMID: 22995223 DOI: 10.1016/j.tips.2012.08.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 08/18/2012] [Accepted: 08/21/2012] [Indexed: 11/23/2022]
Abstract
The Janus kinase (JAK) proteins are a family of intracellular nonreceptor tyrosine kinases involved in cytokine signaling via the JAK-STAT (signal transducers and activators of transcription) pathway. Genetic studies have identified somatic JAK2(V617F) mutations and other mutant alleles that activate JAK-STAT signaling in most patients with myeloproliferative neoplasms (MPNs). As a result, JAK inhibitors have been developed to treat various malignancies and have been shown to be efficacious in both preclinical and clinical settings. However, available ATP-competitive JAK (type I) inhibitors are associated with dose-dependent toxicities, and do not yet reduce disease burden in MPN patients. Recent studies suggest that genetic and epigenetic mechanisms can cause insensitivity to type I JAK inhibitors. Novel therapies include the development of type II JAK inhibitors and the use of alternative strategies to abrogate JAK-STAT signaling, perhaps with histone deacetylase (HDAC) and heat shock protein 90 (HSP90) inhibitors. These innovative therapies may translate to treatment of other diseases that are dependent on JAK signaling, including B-precursor acute lymphoblastic leukemia (B-ALL).
Collapse
|
241
|
Xu LS, Sokalski KM, Hotke K, Christie DA, Zarnett O, Piskorz J, Thillainadesan G, Torchia J, DeKoter RP. Regulation of B Cell Linker Protein Transcription by PU.1 and Spi-B in Murine B Cell Acute Lymphoblastic Leukemia. THE JOURNAL OF IMMUNOLOGY 2012; 189:3347-54. [DOI: 10.4049/jimmunol.1201267] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
242
|
Bains T, Heinrich MC, Loriaux MM, Beadling C, Nelson D, Warrick A, Neff TL, Tyner JW, Dunlap J, Corless CL, Fan G. Newly described activating JAK3 mutations in T-cell acute lymphoblastic leukemia. Leukemia 2012; 26:2144-6. [PMID: 22425895 DOI: 10.1038/leu.2012.74] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
243
|
Zhou Y, You MJ, Young KH, Lin P, Lu G, Medeiros LJ, Bueso-Ramos CE. Advances in the molecular pathobiology of B-lymphoblastic leukemia. Hum Pathol 2012; 43:1347-1362. [PMID: 22575265 DOI: 10.1016/j.humpath.2012.02.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 01/26/2012] [Accepted: 02/02/2012] [Indexed: 02/03/2023]
Abstract
B-lymphoblastic leukemia/lymphoma, also known as B-acute lymphoblastic leukemia, is derived from B-cell progenitors. B-acute lymphoblastic leukemia occurs predominantly in children, but can occur at any age. Risk-adapted intensive chemotherapy is effective in treating most children with B-acute lymphoblastic leukemia, but this approach is less successful in adults. Recent developments in genome-wide genetic analysis in B-acute lymphoblastic leukemia have provided insights into disease pathogenesis and prognosis. B-acute lymphoblastic leukemia cases usually carry a primary genetic event, often a chromosome translocation, and a constellation of secondary genetic alterations that are acquired and selected dynamically in a nonlinear fashion. These genetic changes commonly affect cellular mechanisms that control B-cell differentiation and proliferation. The cooperative interaction between inactivation of hematopoietic transcription factors involved in differentiation (class II mutation) and activating mutations involved in cell proliferation (class I mutation) is reminiscent of the pathogenic model of acute myeloid leukemia. The resulting improved molecular understanding of B-acute lymphoblastic leukemia is helping to refine disease risk stratification and discover new therapeutic approaches for patients with refractory disease. In this review, we first summarize the clinicopathologic and immunophenotypic features of B-acute lymphoblastic leukemia and introduce current understanding of B-cell development and B-acute lymphoblastic leukemia leukemogenesis. We then focus on recent advances in genetic analysis and gene expression profiling of B-acute lymphoblastic leukemia and discuss the implications of these findings for disease evolution, risk prediction, and possible novel therapeutic approaches.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
244
|
Differences in signaling through the B-cell leukemia oncoprotein CRLF2 in response to TSLP and through mutant JAK2. Blood 2012; 120:2853-63. [PMID: 22915648 DOI: 10.1182/blood-2012-02-413252] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Approximately 10% of B-cell acute lymphoblastic leukemias (B-ALLs) overexpress the cytokine receptor subunit CRLF2, which may confer a poor prognosis. CRLF2 binds its ligand thymic stromal lymphopoietin (TSLP) as a heterodimer with IL7R. Subsets of CRLF2-overexpressing B-ALLs also have a gain-of-function CRLF2 F232C mutation or activating mutations in JAK2. Whether these mutant alleles confer differences in signaling has not been addressed. Through a domain mutation analysis, we demonstrate a distinct dependence on the CRLF2 intracellular tyrosine Y368 in signaling by CRLF2 F232C, but not signaling induced by TSLP or through CRLF2/mutant JAK2. In contrast, CRLF2 signaling in each context is strictly dependent on both the CRLF2 box1 domain and the intracellular tryptophan W286. Using a global quantitative analysis of tyrosine phosphorylation induced by TSLP, we previously identified TSLP-induced phosphorylation of multiple kinases implicated in B-cell receptor signaling, including Lyn, Btk, Hck, Syk, MAPK8, MAPK9, and MAPK10. We now demonstrate that cells dependent on CRLF2/mutant JAK2 have reduced phosphorylation at these targets, suggesting that the kinases promote TSLP-mediated proliferation but serve as negative regulators of CRLF2/mutant JAK2 signaling. Thus, targetable nodes downstream of CRLF2 differ based on the presence or absence of additional mutations in CRLF2 signaling components.
Collapse
|
245
|
BET bromodomain inhibition targets both c-Myc and IL7R in high-risk acute lymphoblastic leukemia. Blood 2012; 120:2843-52. [PMID: 22904298 DOI: 10.1182/blood-2012-02-413021] [Citation(s) in RCA: 310] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We investigated the therapeutic potential of JQ1, an inhibitor of the BET class of human bromodomain proteins, in B-cell acute lymphoblastic leukemia (B-ALL). We show that JQ1 potently reduces the viability of B-ALL cell lines with high-risk cytogenetics. Among the most sensitive were lines with rearrangements of CRLF2, which is overexpressed in ~ 10% of B-ALL. CRLF2 heterodimerizes with the IL7 receptor (IL7R) and signals through JAK2, JAK1, and STAT5 to drive proliferation and suppress apoptosis. As previously observed, JQ1 induced the down-regulation of MYC transcription, the loss of BRD4 at the MYC promoter, and the reduced expression of c-Myc target genes. Strikingly, JQ1 also down-regulated IL7R transcription, depleted BRD4 from the IL7R promoter, and reduced JAK2 and STAT5 phosphorylation. Genome-wide expression profiling demonstrated a restricted effect of JQ1 on transcription, with MYC and IL7R being among the most down-regulated genes. Indeed, IL7R was the only cytokine receptor in CRLF2-rearranged B-ALL cells significantly down-regulated by JQ1 treatment. In mice xenografted with primary human CRLF2-rearranged B-ALL, JQ1 suppressed c-Myc expression and STAT5 phosphorylation and significantly prolonged survival. Thus, bromodomain inhibition is a promising therapeutic strategy for B-ALL as well as other conditions dependent on IL7R signaling.
Collapse
|
246
|
Roberts KG, Morin RD, Zhang J, Hirst M, Zhao Y, Su X, Chen SC, Payne-Turner D, Churchman M, Harvey RC, Chen X, Kasap C, Yan C, Becksfort J, Finney RP, Teachey DT, Maude SL, Tse K, Moore R, Jones S, Mungall K, Birol I, Edmonson MN, Hu Y, Buetow KE, Chen IM, Carroll WL, Wei L, Ma J, Kleppe M, Levine RL, Garcia-Manero G, Larsen E, Shah NP, Devidas M, Reaman G, Smith M, Paugh SW, Evans WE, Grupp SA, Jeha S, Pui CH, Gerhard DS, Downing JR, Willman CL, Loh M, Hunger SP, Marra M, Mullighan CG. Genetic alterations activating kinase and cytokine receptor signaling in high-risk acute lymphoblastic leukemia. Cancer Cell 2012; 22:153-66. [PMID: 22897847 PMCID: PMC3422513 DOI: 10.1016/j.ccr.2012.06.005] [Citation(s) in RCA: 528] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 05/21/2012] [Accepted: 06/11/2012] [Indexed: 12/15/2022]
Abstract
Genomic profiling has identified a subtype of high-risk B-progenitor acute lymphoblastic leukemia (B-ALL) with alteration of IKZF1, a gene expression profile similar to BCR-ABL1-positive ALL and poor outcome (Ph-like ALL). The genetic alterations that activate kinase signaling in Ph-like ALL are poorly understood. We performed transcriptome and whole genome sequencing on 15 cases of Ph-like ALL and identified rearrangements involving ABL1, JAK2, PDGFRB, CRLF2, and EPOR, activating mutations of IL7R and FLT3, and deletion of SH2B3, which encodes the JAK2-negative regulator LNK. Importantly, several of these alterations induce transformation that is attenuated with tyrosine kinase inhibitors, suggesting the treatment outcome of these patients may be improved with targeted therapy.
Collapse
Affiliation(s)
- Kathryn G. Roberts
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Ryan D. Morin
- Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3
| | - Jinghui Zhang
- Department of Computational Biology and Bioinformatics, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Martin Hirst
- Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3
| | - Yongjun Zhao
- Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3
| | - Xiaoping Su
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Shann-Ching Chen
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Debbie Payne-Turner
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Michelle Churchman
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Richard C. Harvey
- University of New Mexico Cancer Research and Treatment Center, Albuquerque, NM 87131
| | - Xiang Chen
- Department of Computational Biology and Bioinformatics, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Corynn Kasap
- School of Medicine, University of California, San Francisco, CA 94143
| | - Chunhua Yan
- Center for Bioinformatics and Information Technology, National Institutes of Health, Bethesda, MD 20892
| | - Jared Becksfort
- Department of Information Sciences, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Richard P. Finney
- Center for Bioinformatics and Information Technology, National Institutes of Health, Bethesda, MD 20892
| | - David T. Teachey
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Shannon L. Maude
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Kane Tse
- Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3
| | - Richard Moore
- Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3
| | - Steven Jones
- Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3
| | - Karen Mungall
- Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3
| | - Inanc Birol
- Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3
| | - Michael N. Edmonson
- Laboratory of Population Genetics, National Institutes of Health, Bethesda, MD 20892
| | - Ying Hu
- Laboratory of Population Genetics, National Institutes of Health, Bethesda, MD 20892
| | - Kenneth E. Buetow
- Laboratory of Population Genetics, National Institutes of Health, Bethesda, MD 20892
| | - I-Ming Chen
- University of New Mexico Cancer Research and Treatment Center, Albuquerque, NM 87131
| | | | - Lei Wei
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Jing Ma
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Maria Kleppe
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Ross L. Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | | | - Eric Larsen
- Maine Children’s Cancer Program, Scarborough, ME 04074
| | - Neil P. Shah
- School of Medicine, University of California, San Francisco, CA 94143
| | - Meenakshi Devidas
- Epidemiology and Health Policy Research, University of Florida, Gainesville, FL 32601
| | - Gregory Reaman
- Children’s National Medical Center, Washington, DC 20010
| | - Malcolm Smith
- Office of Cancer Genomics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Steven W. Paugh
- Department of Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - William E. Evans
- Department of Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Stephan A. Grupp
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Sima Jeha
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Ching-Hon Pui
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Daniela S. Gerhard
- Office of Cancer Genomics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - James R. Downing
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN 38105
| | - Cheryl L. Willman
- University of New Mexico Cancer Research and Treatment Center, Albuquerque, NM 87131
| | - Mignon Loh
- Department of Pediatrics, University of California, San Francisco, CA 94143
| | - Stephen P. Hunger
- University of Colorado School of Medicine and The Children’s Hospital Colorado, Aurora, CO 80045
| | - Marco Marra
- Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 1L3
- Department of Medical Genetics, University of British Columbia, Vancouver, BC VSZ 1L3
| | - Charles G. Mullighan
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN 38105
| |
Collapse
|
247
|
|
248
|
Chen E, Staudt LM, Green AR. Janus kinase deregulation in leukemia and lymphoma. Immunity 2012; 36:529-41. [PMID: 22520846 DOI: 10.1016/j.immuni.2012.03.017] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Indexed: 12/21/2022]
Abstract
Genetic alterations affecting members of the Janus kinase (JAK) family have been discovered in a wide array of cancers and are particularly prominent in hematological malignancies. In this review, we focus on the role of such lesions in both myeloid and lymphoid tumors. Oncogenic JAK molecules can activate a myriad of canonical downstream signaling pathways as well as directly interact with chromatin in noncanonical processes, the interplay of which results in a plethora of diverse biological consequences. Deciphering these complexities is shedding unexpected light on fundamental cellular mechanisms and will also be important for improved diagnosis, identification of new therapeutic targets, and the development of stratified approaches to therapy.
Collapse
Affiliation(s)
- Edwin Chen
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
249
|
Aberrant STAT5 and PI3K/mTOR pathway signaling occurs in human CRLF2-rearranged B-precursor acute lymphoblastic leukemia. Blood 2012; 120:833-42. [PMID: 22685175 DOI: 10.1182/blood-2011-12-389932] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adults and children with high-risk CRLF2-rearranged acute lymphoblastic leukemia (ALL) respond poorly to current cytotoxic chemotherapy and suffer unacceptably high rates of relapse, supporting the need to use alternative therapies. CRLF2 encodes the thymic stromal lymphopoietin (TSLP) receptor, which activates cell signaling in normal lymphocytes on binding its ligand, TSLP. We hypothesized that aberrant cell signaling occurs in CRLF2-rearranged ALL and can be targeted by signal transduction inhibitors of this pathway. In a large number of primary CRLF2-rearranged ALL samples, we observed increased basal levels of pJAK2, pSTAT5, and pS6. We thus characterized the biochemical sequelae of CRLF2 and JAK alterations in CRLF2-rearranged ALL primary patient samples via analysis of TSLP-mediated signal transduction. TSLP stimulation of these leukemias further induced robust JAK/STAT and PI3K/mTOR pathway signaling. JAK inhibition abrogated phosphorylation of JAK/STAT and, surprisingly, of PI3K/mTOR pathway members, suggesting an interconnection between these signaling networks and providing a rationale for testing JAK inhibitors in clinical trials. The PI3K/mTOR pathway inhibitors rapamycin, PI103, and PP242 also inhibited activated signal transduction and translational machinery proteins of the PI3K/mTOR pathway, suggesting that signal transduction inhibitors targeting this pathway also may have therapeutic relevance for patients with CRLF2-rearranged ALL and merit further preclinical testing.
Collapse
|
250
|
Abstract
IL-7 plays many essential roles in human health and disease. Congenital deficiencies in IL-7 signaling result in profound immunodeficiency, polymorphisms in IL7Rα modulate susceptibility to autoimmune disease, and acquired somatic activating mutations in IL7Rα contribute to neoplastic transformation in B cell and T cell leukemia. In response to lymphopenia, IL-7 accumulates to supranormal levels, which alters T cell homeostasis by augmenting T cell reactivity toward self and cognate antigens. This physiologic response is now routinely exploited to improve the efficacy of adoptive cell therapies for cancer. Clinical trials of recombinant IL-7 have demonstrated safety and potent immunorestorative effects, and current studies are investigating whether rhIL-7 therapy can improve outcomes in chronic viral infection and in the context of cancer immunotherapies. Building upon the large fund of knowledge regarding the basic biology of IL-7, this review will discuss the many and varied roles of IL-7 in human health and disease.
Collapse
Affiliation(s)
- Wangko Lundström
- Pediatric Oncology Branch, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, United States.
| | | | | |
Collapse
|