1
|
Guo YJ, Ma MX, Tian T, Zhang JN, Guo XN, Qiao S. Myeloid/lymphoid neoplasms with eosinophilia and FGFR1 rearrangement t(8;13)(p11;q12): A case report and literature review. Oncol Lett 2024; 28:468. [PMID: 39119236 PMCID: PMC11306990 DOI: 10.3892/ol.2024.14601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/25/2024] [Indexed: 08/10/2024] Open
Abstract
8p11 myeloproliferative syndrome (EMS) is a rare and aggressive hematological malignancy, characterized by myeloproliferative neoplasms, and associated with eosinophilia and T- or B-cell lineage lymphoblastic lymphoma. The pathogenesis is defined by the presence of chromosomal translocations associated with the fibroblast growth factor-1 (FGFR1) gene, located in the 8p11-12.1 chromosomal locus. At present, only ~100 cases have been reported globally. At least 15 partner genes have been identified, including the most common, the zinc finger MYM-type containing 2 (ZNF198)-FGFR1 fusion gene formed by t(8;13)(p11;q12). Different fusion genes determine the clinical manifestations and prognosis of the disease. Patients with EMS with t(8;13)(p11;q12) commonly present with lymphadenopathy and T-lymphoblastic lymphoma, which usually converts to acute myeloid leukemia (AML) with the progression of the disease. The present study describes the case of an elderly female patient with EMS with t(8;13)(p11;q12), presenting with myeloid/lymphoid syndrome (myeloproliferative neoplasms and T lymphoblastic lymphoma). The patient received the CHOPE regimen combined with tyrosine kinase inhibitor (dasatin) treatment and obtained short-term complete remission. However, 6 months later, the disease progressed from EMS to AML and the patient died due to ineffective induction therapy. The present study also reviews the relevant literature about this unusual entity to enhance the understanding of EMS.
Collapse
Affiliation(s)
- Yu-Jie Guo
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Meng-Xue Ma
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Tian Tian
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jing-Nan Zhang
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiao-Nan Guo
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Shukai Qiao
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
2
|
Shomali W, Gotlib J. World Health Organization and International Consensus Classification of eosinophilic disorders: 2024 update on diagnosis, risk stratification, and management. Am J Hematol 2024; 99:946-968. [PMID: 38551368 DOI: 10.1002/ajh.27287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 04/09/2024]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of non-hematologic (secondary or reactive) and hematologic (primary or clonal) disorders with the potential for end-organ damage. DIAGNOSIS Hypereosinophilia (HE) has generally been defined as a peripheral blood eosinophil count greater than 1.5 × 109/L, and may be associated with tissue damage. After the exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on a combination of various tests. They include morphologic review of the blood and marrow, standard cytogenetics, fluorescence in situ hybridization, molecular testing and flow immunophenotyping to detect histopathologic or clonal evidence for an acute or chronic hematolymphoid neoplasm. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2022 World Health Organization and International Consensus Classification endorse a semi-molecular classification scheme of disease subtypes. This includes the major category "myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions" (MLN-eo-TK), and the MPN subtype, "chronic eosinophilic leukemia" (CEL). Lymphocyte-variant HE is an aberrant T-cell clone-driven reactive eosinophila, and idiopathic hypereosinophilic syndrome (HES) is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (e.g., <1.5 × 109/L) without symptoms or signs of organ involvement, a watch and wait approach with close follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Pemigatinib was recently approved for patients with relapsed or refractory FGFR1-rearranged neoplasms. Corticosteroids are first-line therapy for patients with lymphocyte-variant HE and HES. Hydroxyurea and interferon-α have demonstrated efficacy as initial treatment and in steroid-refractory cases of HES. Mepolizumab, an interleukin-5 (IL-5) antagonist monoclonal antibody, is approved by the U.S Food and Drug Administration for patients with idiopathic HES. Cytotoxic chemotherapy agents, and hematopoietic stem cell transplantation have been used for aggressive forms of HES and CEL, with outcomes reported for limited numbers of patients. Targeted therapies such as the IL-5 receptor antibody benralizumab, IL-5 monoclonal antibody depemokimab, and various tyrosine kinase inhibitors for MLN-eo-TK, are under active investigation.
Collapse
Affiliation(s)
- William Shomali
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, California, USA
| | - Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
3
|
Morales-Camacho RM, Caballero-Velázquez T, Borrero JJ, Bernal R, Prats-Martín C. Hematological Neoplasms with Eosinophilia. Cancers (Basel) 2024; 16:337. [PMID: 38254826 PMCID: PMC10814743 DOI: 10.3390/cancers16020337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Eosinophils in peripheral blood account for 0.3-5% of leukocytes, which is equivalent to 0.05-0.5 × 109/L. A count above 0.5 × 109/L is considered to indicate eosinophilia, while a count equal to or above 1.5 × 109/L is defined as hypereosinophilia. In bone marrow aspirate, eosinophilia is considered when eosinophils make up more than 6% of the total nuclear cells. In daily clinical practice, the most common causes of reactive eosinophilia are non-hematologic, whether they are non-neoplastic (allergic diseases, drugs, infections, or immunological diseases) or neoplastic (solid tumors). Eosinophilia that is associated with a hematological malignancy may be reactive or secondary to the production of eosinophilopoietic cytokines, and this is mainly seen in lymphoid neoplasms (Hodgkin lymphoma, mature T-cell neoplasms, lymphocytic variant of hypereosinophilic syndrome, and B-acute lymphoblastic leukemia/lymphoma). Eosinophilia that is associated with a hematological malignancy may also be neoplastic or primary, derived from the malignant clone, usually in myeloid neoplasms or with its origin in stem cells (myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions, acute myeloid leukemia with core binding factor translocations, mastocytosis, myeloproliferative neoplasms, myelodysplastic/myeloproliferative neoplasms, and myelodysplastic neoplasms). There are no concrete data in standardized cytological and cytometric procedures that could predict whether eosinophilia is reactive or clonal. The verification is usually indirect, based on the categorization of the accompanying hematologic malignancy. This review focuses on the broad differential diagnosis of hematological malignancies with eosinophilia.
Collapse
Affiliation(s)
- Rosario M. Morales-Camacho
- Department of Hematology, Virgen del Rocío University Hospital, Seville Biomedicine Institute (IBiS/CSIC), University of Seville, 41013 Seville, Spain (R.B.)
| | - Teresa Caballero-Velázquez
- Department of Hematology, Virgen del Rocío University Hospital, Seville Biomedicine Institute (IBiS/CSIC), University of Seville, 41013 Seville, Spain (R.B.)
| | - Juan José Borrero
- Department of Pathology, Virgen del Rocío University Hospital, 41013 Seville, Spain;
| | - Ricardo Bernal
- Department of Hematology, Virgen del Rocío University Hospital, Seville Biomedicine Institute (IBiS/CSIC), University of Seville, 41013 Seville, Spain (R.B.)
| | - Concepción Prats-Martín
- Department of Hematology, Virgen del Rocío University Hospital, Seville Biomedicine Institute (IBiS/CSIC), University of Seville, 41013 Seville, Spain (R.B.)
| |
Collapse
|
4
|
Shomali W, Colucci P, George TI, Kiladjian JJ, Langford C, Patel JL, Reiter A, Vannucchi AM, Gotlib J. Comprehensive response criteria for myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions: a proposal from the MLN International Working Group. Leukemia 2023; 37:981-987. [PMID: 37076693 PMCID: PMC10169632 DOI: 10.1038/s41375-023-01859-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/20/2022] [Accepted: 02/21/2023] [Indexed: 04/21/2023]
Affiliation(s)
- William Shomali
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA
| | | | - Tracy I George
- ARUP Laboratories and University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Cheryl Langford
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA
| | - Jay L Patel
- ARUP Laboratories and University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | | | - Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
Freyer CW, Hughes ME, Carulli A, Bagg A, Hexner E. Pemigatinib for the treatment of myeloid/lymphoid neoplasms with FGFR1 rearrangement. Expert Rev Anticancer Ther 2023; 23:351-359. [PMID: 36927350 DOI: 10.1080/14737140.2023.2192930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
INTRODUCTION Myeloid/lymphoid neoplasms with fibroblast growth factor receptor-1 (FGFR1) rearrangements (MLNFGFR1) are rare entities with aggressive features and poor prognosis. Presentation is heterogeneous, ranging from myeloproliferative neoplasms (with or without eosinophilia) to T-cell lymphoma and acute leukemia. Historical treatments have been guided by the presenting phenotype with induction chemotherapy frequently used. Pemigatinib is a FGFR1-3 tyrosine kinase inhibitor that has demonstrated high complete hematologic and cytogenetic response rates in MLNFGFR1. AREAS COVERED We discuss the pathogenesis, presentation, and historical treatments for MLNFGFR1, in addition to clinical data using pemigatinib and other targeted therapies. Discussion of the mechanism of action and adverse events are also included. EXPERT OPINION Pemigatinib represents a significant advance in the management of MLNFGFR1. High rates of complete hematologic and cytogenetic response have been observed. While direct comparative data are unavailable, outcomes appear favorable compared to conventional approaches. Long term efficacy and tolerability are not yet known, and allogeneic hematopoietic stem cell transplant (alloHSCT) has been and continues to be the treatment with the highest chance of long term disease free survival in responding patients. Combinations of pemigatinib and chemotherapy, particularly for more aggressive phenotypes, warrant future investigation as does the use of pemigatinib maintenance following alloHSCT.
Collapse
Affiliation(s)
- Craig W Freyer
- Department of Pharmacy Services, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.,Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine and the Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
| | - Mitchell E Hughes
- Department of Pharmacy Services, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.,Lymphoma Program, Hematology/Oncology Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alison Carulli
- Department of Pharmacy Services, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Hexner
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine and the Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
6
|
Biologic therapies for hypereosinophilic disorders: From tyrosine kinase inhibitors to monoclonal antibodies. Towards an increasingly customized management? Blood Rev 2023; 58:101014. [PMID: 36153195 DOI: 10.1016/j.blre.2022.101014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/10/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022]
Abstract
Hypereosinophilic syndromes (HES) encompass a wide range of disorders characterized by persistent peripheral blood hypereosinophilia (HE) (i.e., an eosinophil count ≥1.5 × 109/L and ≥ 10% eosinophils preferably with a minimal duration of 6 months if documentation is available) associated with organ damage and/or dysfunction attributable to tissue eosinophilic infiltrate and release of granule contents. In most cases, HE is associated with atopic conditions/allergies, parasitic infections, medications, autoimmune disorders and/or solid tumors in most cases. More rarely, it can be one of the dominant manifestations of an underlying myeloid/lymphoid neoplasm. With regard to hematological forms, in recent decades the advances in understanding the pathogenic aspects of HES have led to a growing interest in these diseases, and in the 2016 WHO classification multiple subgroups were defined according to the molecular profile with the aim of better characterizing these syndromes and establishing which patients will benefit from specific pharmacological targeted therapies. This review article will provide a comprehensive overview of possible therapeutic approaches for HES in the light of each specific molecular alteration, considering both tyrosine kinase inhibitors and monoclonal antibodies, either implemented in clinical practice or currently still under development.
Collapse
|
7
|
Gotlib J. Available and emerging therapies for bona fide advanced systemic mastocytosis and primary eosinophilic neoplasms. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:34-46. [PMID: 36485158 PMCID: PMC9821059 DOI: 10.1182/hematology.2022000368] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The historically poor prognosis of patients with advanced systemic mastocytosis (AdvSM) and primary eosinophilic neoplasms has shifted to increasingly favorable outcomes with the discovery of druggable targets. The multikinase/KIT inhibitor midostaurin and the highly selective KIT D816V inhibitor avapritinib can elicit marked improvements in measures of mast cell (MC) burden as well as reversion of MC-mediated organ damage (C-findings) and disease symptoms. With avapritinib, the achievement of molecular remission of KIT D816V and improved survival compared with historical therapy suggests a potential to affect disease natural history. BLU-263 and bezuclastinib are KIT D816V inhibitors currently being tested in trials of AdvSM. In the new World Health Organization and International Consensus Classifications, the category of "myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase (TK) gene fusions" is inclusive of rearrangements involving PDGFRA, PDGFRB, FGFR1, JAK2, FLT3, and ETV6::ABL1. While the successful outcomes with imatinib in FIP1L1::PDGFRA-positive cases and PDGFRB-rearranged neoplasms have become the "poster children" of these disorders, the responses of the other TK-driven neoplasms to small-molecule inhibitors are more variable. The selective FGFR inhibitor pemigatinib, approved in August 2022, is a promising therapy in aggressive FGFR1-driven diseases and highlights the role of such agents in bridging patients to allogeneic transplantation. This review summarizes the data for these approved and investigational agents and discusses open questions and future priorities regarding the management of these rare diseases.
Collapse
Affiliation(s)
- Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
8
|
Li T, Zhang G, Zhang X, Lin H, Liu Q. The 8p11 myeloproliferative syndrome: Genotypic and phenotypic classification and targeted therapy. Front Oncol 2022; 12:1015792. [PMID: 36408177 PMCID: PMC9669583 DOI: 10.3389/fonc.2022.1015792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/10/2022] [Indexed: 10/05/2023] Open
Abstract
EMS(8p11 myeloproliferative syndrome, EMS) is an aggressive hematological neoplasm with/without eosinophilia caused by a rearrangement of the FGFR1 gene at 8p11-12. It was found that all cases carry chromosome abnormalities at the molecular level, not only the previously reported chromosome translocation and insertion but also a chromosome inversion. These abnormalities produced 17 FGFR1 fusion genes, of which the most common partner genes are ZNF198 on 13q11-12 and BCR of 22q11.2. The clinical manifestations can develop into AML (acute myeloid leukemia), T-LBL (T-cell lymphoblastic lymphoma), CML (chronic myeloid leukemia), CMML (chronic monomyelocytic leukemia), or mixed phenotype acute leukemia (MPAL). Most patients are resistant to traditional chemotherapy, and a minority of patients achieve long-term clinical remission after stem cell transplantation. Recently, the therapeutic effect of targeted tyrosine kinase inhibitors (such as pemigatinib and infigratinib) in 8p11 has been confirmed in vitro and clinical trials. The TKIs may become an 8p11 treatment option as an alternative to hematopoietic stem cell transplantation, which is worthy of further study.
Collapse
Affiliation(s)
- Taotao Li
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Gaoling Zhang
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Hai Lin
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Qiuju Liu
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
9
|
Shomali W, Gotlib J. World Health Organization-defined eosinophilic disorders: 2022 update on diagnosis, risk stratification, and management. Am J Hematol 2022; 97:129-148. [PMID: 34533850 DOI: 10.1002/ajh.26352] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of nonhematologic (secondary or reactive) and hematologic (primary or clonal) disorders with potential for end-organ damage. DIAGNOSIS Hypereosinophilia (HE) has generally been defined as a peripheral blood eosinophil count greater than 1.5 × 109 /L. After exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on morphologic review of the blood and marrow, standard cytogenetics, fluorescence in situ hybridization, next generation sequencing gene assays, and flow immunophenotyping to detect histopathologic or clonal evidence for an acute or chronic hematolymphoid neoplasm. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2016 World Health Organization endorses a semi-molecular classification scheme of disease subtypes. This includes the major category "myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB, or FGFR1 or with PCM1-JAK2", and the myeloproliferative neoplasm subtype, "chronic eosinophilic leukemia, not otherwise specified" (CEL, NOS). Lymphocyte-variant HE is an aberrant T-cell clone-driven reactive eosinophila, and idiopathic hypereosinophilic syndrome (HES) is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (eg, < 1.5 × 109 /L) without symptoms or signs of organ involvement, a watch and wait approach with close follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Corticosteroids are first-line therapy for patients with lymphocyte-variant HE and HES. Hydroxyurea and interferon-α have demonstrated efficacy as initial treatment and in steroid-refractory cases of HES. Mepolizumab, an interleukin-5 (IL-5) antagonist monoclonal antibody, was recently approved by the US Food and Drug Administration for patients with idiopathic HES. The use of the IL-5 receptor antibody benralizumab, as well as other targeted therapies such as JAK2 and FGFR1 inhibitors, is under active investigation.
Collapse
Affiliation(s)
- William Shomali
- Division of Hematology, Stanford Cancer Institute Stanford University School of Medicine Stanford California USA
| | - Jason Gotlib
- Division of Hematology, Stanford Cancer Institute Stanford University School of Medicine Stanford California USA
| |
Collapse
|
10
|
Cowell JK, Hu T. Mechanisms of resistance to FGFR1 inhibitors in FGFR1-driven leukemias and lymphomas: implications for optimized treatment. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:607-619. [PMID: 34734169 PMCID: PMC8562765 DOI: 10.20517/cdr.2021.30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myeloid and lymphoid neoplasms with eosinophilia and FGFR1 rearrangements (MLN-eo FGFR1) disease is derived from a pluripotent hematopoietic stem cell and has a complex presentation with a myeloproliferative disorder with or without eosinophilia and frequently presents with mixed lineage T- or B-lymphomas. The myeloproliferative disease frequently progresses to AML and lymphoid neoplasms can develop into acute lymphomas. No matter the cell type involved, or clinical presentation, chromosome translocations involving the FGFR1 kinase and various partner genes, which leads to constitutive activation of downstream oncogenic signaling cascades. These patients are not responsive to treatment regimens developed for other acute leukemias and survival is poor. Recent development of specific FGFR1 inhibitors has suggested an alternative therapeutic approach but resistance is likely to evolve over time. Mouse models of this disease syndrome have been developed and are being used for preclinical evaluation of FGFR1 inhibitors. Cell lines from these models have now been developed and have been used to investigate the mechanisms of resistance that might be expected in clinical cases. So far, a V561M mutation in the kinases domain and deletion of PTEN have been recognized as leading to resistance and both operate through the PI3K/AKT signaling axis. One of the important consequences is the suppression of PUMA, a potent enforcer of apoptosis, which operates through BCL2. Targeting BCL2 in the resistant cells leads to suppression of leukemia development in mouse models, which potentially provides an opportunity to treat patients that become resistant to FGFR1 inhibitors. In addition, elucidation of molecular mechanisms underlying FGFR1-driven leukemias and lymphomas also provides new targets for combined treatment as another option to bypass the FGFR1 inhibitor resistance and improve patient outcome.
Collapse
Affiliation(s)
- John K Cowell
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Tianxiang Hu
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
| |
Collapse
|
11
|
Chen X, Wang F, Zhang Y, Ma X, Cao P, Yuan L, Wang L, Chen J, Zhou X, Wu Q, Liu M, Jin D, Liu H. Fusion gene map of acute leukemia revealed by transcriptome sequencing of a consecutive cohort of 1000 cases in a single center. Blood Cancer J 2021; 11:112. [PMID: 34135310 PMCID: PMC8209121 DOI: 10.1038/s41408-021-00504-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/15/2021] [Accepted: 05/26/2021] [Indexed: 12/28/2022] Open
Abstract
Fusion genes (FGs) are important genetic abnormalities in acute leukemias, but their variety and occurrence in acute leukemias remain to be systematically described. Whole transcriptome sequencing (WTS) provides a powerful tool for analyzing FGs. Here we report the FG map revealed by WTS in a consecutive cohort of 1000 acute leukemia cases in a single center, including 539 acute myeloid leukemia (AML), 437 acute lymphoblastic leukemia (ALL), and 24 mixed-phenotype acute leukemia (MPAL) patients. Bioinformatic analysis identified 792 high-confidence in-frame fusion events (296 distinct fusions) which were classified into four tiers. Tier A (pathogenic), B (likely pathogenic), and C (uncertain significance) FGs were identified in 61.8% cases of the total cohort (59.7% in AML, 64.5% in ALL, and 63.6% in MPAL). FGs involving protein kinase, transcription factor, and epigenetic genes were detected in 10.7%, 48.5%, and 15.1% cases, respectively. A considerable amount of novel FGs (82 in AML, 88 in B-ALL, 13 in T-ALL, and 9 in MPAL) was identified. This comprehensively described real map of FGs in acute leukemia revealed multiple FGs with clinical relevance that have not been previously recognized. WTS is a valuable tool and should be widely used in the routine diagnostic workup of acute leukemia.
Collapse
Affiliation(s)
- Xue Chen
- Division of Pathology & Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, 065201, Langfang, China
| | - Fang Wang
- Division of Pathology & Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, 065201, Langfang, China
| | - Yang Zhang
- Division of Pathology & Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, 065201, Langfang, China
| | - Xiaoli Ma
- Division of Pathology & Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, 065201, Langfang, China
| | - Panxiang Cao
- Division of Pathology & Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, 065201, Langfang, China
| | - Lili Yuan
- Division of Pathology & Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, 065201, Langfang, China
| | - Lan Wang
- Division of Pathology & Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, 065201, Langfang, China
| | - Jiaqi Chen
- Division of Pathology & Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, 065201, Langfang, China
| | - Xiaosu Zhou
- Beijing Lu Daopei Institute of Hematology, 100176, Beijing, China
| | - Qisheng Wu
- Division of Pathology & Laboratory Medicine, Beijing Lu Daopei Hospital, 100176, Beijing, China
| | - Ming Liu
- Division of Pathology & Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, 065201, Langfang, China
| | - David Jin
- Beijing Lu Daopei Institute of Hematology, 100176, Beijing, China
| | - Hongxing Liu
- Division of Pathology & Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, 065201, Langfang, China. .,Beijing Lu Daopei Institute of Hematology, 100176, Beijing, China. .,Division of Pathology & Laboratory Medicine, Beijing Lu Daopei Hospital, 100176, Beijing, China.
| |
Collapse
|
12
|
Renneville A, Gasser JA, Grinshpun DE, Jean Beltran PM, Udeshi ND, Matyskiela ME, Clayton T, McConkey M, Viswanathan K, Tepper A, Guirguis AA, Sellar RS, Cotteret S, Marzac C, Saada V, De Botton S, Kiladjian JJ, Cayuela JM, Rolfe M, Chamberlain PP, Carr SA, Ebert BL. Avadomide induces degradation of ZMYM2 fusion oncoproteins in hematologic malignancies. Blood Cancer Discov 2021; 2:250-265. [PMID: 34027417 DOI: 10.1158/2643-3230.bcd-20-0105] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Thalidomide analogs exert their therapeutic effects by binding to the CRL4CRBN E3 ubiquitin ligase, promoting ubiquitination and subsequent proteasomal degradation of specific protein substrates. Drug-induced degradation of IKZF1 and IKZF3 in B-cell malignancies demonstrates the clinical utility of targeting disease-relevant transcription factors for degradation. Here, we found that avadomide (CC-122) induces CRBN-dependent ubiquitination and proteasomal degradation of ZMYM2 (ZNF198), a transcription factor involved in balanced chromosomal rearrangements with FGFR1 and FLT3 in aggressive forms of hematologic malignancies. The minimal drug-responsive element of ZMYM2 is a zinc-chelating MYM domain and is contained in the N-terminal portion of ZMYM2 that is universally included in the derived fusion proteins. We demonstrate that avadomide has the ability to induce proteasomal degradation of ZMYM2-FGFR1 and ZMYM2-FLT3 chimeric oncoproteins, both in vitro and in vivo. Our findings suggest that patients with hematologic malignancies harboring these ZMYM2 fusion proteins may benefit from avadomide treatment.
Collapse
Affiliation(s)
- Aline Renneville
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jessica A Gasser
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Daniel E Grinshpun
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Namrata D Udeshi
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Mary E Matyskiela
- Celgene/Bristol-Myers Squibb corporation, San Diego, California, USA
| | - Thomas Clayton
- Celgene/Bristol-Myers Squibb corporation, San Diego, California, USA
| | - Marie McConkey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kaushik Viswanathan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Alexander Tepper
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Andrew A Guirguis
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rob S Sellar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Haematology, UCL Cancer Institute, London, United Kingdom
| | - Sophie Cotteret
- Département de Biologie et Pathologie, Gustave Roussy Cancer Campus, Villejuif, France
| | - Christophe Marzac
- Département de Biologie et Pathologie, Gustave Roussy Cancer Campus, Villejuif, France
| | - Véronique Saada
- Département de Biologie et Pathologie, Gustave Roussy Cancer Campus, Villejuif, France
| | - Stéphane De Botton
- Département d'Hématologie, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jean-Jacques Kiladjian
- Université de Paris, AP-HP, Hôpital Saint-Louis, Centre d'Investigations Cliniques CIC 1427, INSERM, Paris, France
| | - Jean-Michel Cayuela
- Hematology Laboratory and EA3518, University Hospital Saint-Louis, Université de Paris, Paris, France
| | - Mark Rolfe
- Celgene/Bristol-Myers Squibb corporation, San Diego, California, USA
| | | | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Benjamin L Ebert
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Howard Hughes Medical Institute, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Helbig G, Klion AD. Hypereosinophilic syndromes - An enigmatic group of disorders with an intriguing clinical spectrum and challenging treatment. Blood Rev 2021; 49:100809. [PMID: 33714638 DOI: 10.1016/j.blre.2021.100809] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/29/2020] [Accepted: 02/19/2021] [Indexed: 01/16/2023]
Abstract
Hypereosinophilic syndromes (HES) comprises a group of rare disorders characterized by blood hypereosinophilia (>1.5 × 109/l) accompanied by eosinophil-associated organ damage. The 2016 World Health Organization classification recognizes a category of myeloid/lymphoid neoplasms with prominent eosinophilia (M/Leo) and well-characterized gene rearrangements of PDGFRA/B, FGFR1 or JAK2. PDGFRA/B-rearranged patients usually manifest as imatinib-sensitive myeloproliferative neoplasms (MPNs). FGFR1- and JAK2- rearranged cases may manifest as MPNs or aggressive lymphomas/leukemias and historically have had a dismal prognosis, although clinical trials with targeted treatment are promising. A negative screen for M/Leo in a patient with myeloid features should prompt consideration of a diagnosis of chronic eosinophilic leukemia-not otherwise specified. If these are excluded and a secondary cause is not identified, a diagnosis of idiopathic HES and/or other rare variants of HES should be considered. This review, through an illustrative case, summarizes current knowledge on HES pointing at new directions in diagnosis and treatment.
Collapse
Affiliation(s)
- Grzegorz Helbig
- Department of Hematology and Bone Marrow Transplantation, Medical School of Silesia, Silesian Medical University, Katowice, Poland.
| | - Amy D Klion
- Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Gerds AT, Gotlib J, Bose P, Deininger MW, Dunbar A, Elshoury A, George TI, Gojo I, Gundabolu K, Hexner E, Hobbs G, Jain T, Jamieson C, Kuykendall AT, McMahon B, Mohan SR, Oehler V, Oh S, Pardanani A, Podoltsev N, Ranheim E, Rein L, Salit R, Snyder DS, Stein BL, Talpaz M, Thota S, Vachhani P, Wadleigh M, Walsh K, Ward DC, Bergman MA, Sundar H. Myeloid/Lymphoid Neoplasms with Eosinophilia and TK Fusion Genes, Version 3.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020; 18:1248-1269. [PMID: 32886902 DOI: 10.6004/jnccn.2020.0042] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Eosinophilic disorders and related syndromes represent a heterogeneous group of neoplastic and nonneoplastic conditions, characterized by more eosinophils in the peripheral blood, and may involve eosinophil-induced organ damage. In the WHO classification of myeloid and lymphoid neoplasms, eosinophilic disorders characterized by dysregulated tyrosine kinase (TK) fusion genes are recognized as a new category termed, myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB or FGFR1 or with PCM1-JAK2. In addition to these aforementioned TK fusion genes, rearrangements involving FLT3 and ABL1 genes have also been described. These new NCCN Guidelines include recommendations for the diagnosis, staging, and treatment of any one of the myeloid/lymphoid neoplasms with eosinophilia (MLN-Eo) and a TK fusion gene included in the 2017 WHO Classification, as well as MLN-Eo and a FLT3 or ABL1 rearrangement.
Collapse
Affiliation(s)
- Aaron T Gerds
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | | | | | | | - Ivana Gojo
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | - Tania Jain
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | - Vivian Oehler
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | - Stephen Oh
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | | - Rachel Salit
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Brady L Stein
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | - Katherine Walsh
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Dawn C Ward
- UCLA Jonsson Comprehensive Cancer Center; and
| | | | | |
Collapse
|
15
|
Hashemi P, Sadowski I. Diversity of small molecule HIV-1 latency reversing agents identified in low- and high-throughput small molecule screens. Med Res Rev 2020; 40:881-908. [PMID: 31608481 PMCID: PMC7216841 DOI: 10.1002/med.21638] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/26/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
The latency phenomenon produced by human immunodeficiency virus (HIV-1) prevents viral clearance by current therapies, and consequently development of a cure for HIV-1 disease represents a formidable challenge. Research over the past decade has resulted in identification of small molecules that are capable of exposing HIV-1 latent reservoirs, by reactivation of viral transcription, which is intended to render these infected cells sensitive to elimination by immune defense recognition or apoptosis. Molecules with this capability, known as latency-reversing agents (LRAs) could lead to realization of proposed HIV-1 cure strategies collectively termed "shock and kill," which are intended to eliminate the latently infected population by forced reactivation of virus replication in combination with additional interventions that enhance killing by the immune system or virus-mediated apoptosis. Here, we review efforts to discover novel LRAs via low- and high-throughput small molecule screens, and summarize characteristics and biochemical properties of chemical structures with this activity. We expect this analysis will provide insight toward further research into optimized designs for new classes of more potent LRAs.
Collapse
Affiliation(s)
- Pargol Hashemi
- Biochemistry and Molecular Biology, Molecular Epigenetics, Life Sciences InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ivan Sadowski
- Biochemistry and Molecular Biology, Molecular Epigenetics, Life Sciences InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
16
|
Liu YT, Zhao JW, Feng J, Li QH, Chen YM, Qiu LG, Xiao ZJ, Li Y, Gong BF, Gong XY, Mi YC, Wang JX. [Myeloid/lymphoid neoplasms with eosinophilia and FGFR1 rearrangement: 5 cases report and literatures review]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:848-852. [PMID: 31775485 PMCID: PMC7364987 DOI: 10.3760/cma.j.issn.0253-2727.2019.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
目的 分析罕见疾病伴嗜酸性粒细胞增多和FGFR1重排的髓系/淋系肿瘤(即8p11骨髓增殖综合征,EMS)的临床特征、诊断及治疗。 方法 总结中国医学科学院血液病医院2014年1月至2018年5月收治的5例确诊EMS患者的临床表现、实验室特征、诊治经过及转归。 结果 5例EMS患者外周血白细胞计数均明显升高,伴有嗜酸性粒细胞绝对值增高(均值18.89×109/L);骨髓髓系极度增生,原始细胞均<5%,嗜酸性粒细胞比例增高(均值17.24%)。5例患者染色体核型各不相同,但FISH检查均存在FGFR1基因重排。发病至确诊平均时间为4.8个月,中位生存期仅14个月。 结论 EMS是一种罕见病,恶性程度高,对常规化疗反应差,生存期短,且易发生误诊漏诊,细胞遗传学及分子生物学检查有助于早期诊断。
Collapse
Affiliation(s)
- Y T Liu
- Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Blood Diseases, Tianjin 300020, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Iurlo A, Cattaneo D, Gianelli U. Hypereosinophilic syndromes in the precision medicine era: clinical, molecular aspects and therapeutic approaches (targeted therapies). Expert Rev Hematol 2019; 12:1077-1088. [PMID: 31588817 DOI: 10.1080/17474086.2019.1677461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022]
Abstract
Introduction: Hypereosinophilic syndromes are a heterogeneous group of disorders that may be associated with life-threatening organ injury as a result of tissues infiltration by eosinophils. The main goal of therapy is to mitigate eosinophil-mediated organ damage. When possible, therapy should be directed at the underlying etiology. However, even in the absence of any known cause, when organ damage is present, hypereosinophilia must be treated promptly and aggressively to reduce potential morbidity and mortality.Areas covered: Conventional therapies, including corticosteroids, hydroxyurea (hydroxycarbamide) and interferon-alpha, have shown variable efficacy and a non-negligible toxicity emphasizing the need of new therapeutic strategies based on drugs with different mechanisms of action.Expert opinion: Tyrosine kinase inhibitors have a central role among targeted therapies of hypereosinophilic syndromes. Imatinib, initially empirically used based on its activity in chronic myeloid leukemia, achieved preliminary excellent results further confirmed in large series of patients. Third-generation tyrosine kinase inhibitors such as ponatinib, while active in vitro and in vivo in animals, still deserve confirmation in properly designed clinical trials. In addition, clinical investigation on monoclonal antibodies against interleukin-5, interleukin-5Rα, IgE, and CD52 represents a promising area of research.
Collapse
Affiliation(s)
- Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, and University of Milan, Milan, Italy
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, and University of Milan, Milan, Italy
| | - Umberto Gianelli
- Division of Pathology, Department of Pathophysiology and Transplantation, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, and University of Milan, Milan, Italy
| |
Collapse
|
18
|
Brown LM, Bartolo RC, Davidson NM, Schmidt B, Brooks I, Challis J, Petrovic V, Khuong-Quang DA, Mechinaud F, Khaw SL, Majewski IJ, Oshlack A, Ekert PG. Targeted therapy and disease monitoring in CNTRL-FGFR1-driven leukaemia. Pediatr Blood Cancer 2019; 66:e27897. [PMID: 31250523 DOI: 10.1002/pbc.27897] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/13/2019] [Accepted: 06/09/2019] [Indexed: 12/30/2022]
Abstract
We report two patients with leukaemia driven by the rare CNTRL-FGFR1 fusion oncogene. This fusion arises from a t(8;9)(p12;q33) translocation, and is a rare driver of biphenotypic leukaemia in children. We used RNA sequencing to report novel features of expressed CNTRL-FGFR1, including CNTRL-FGFR1 fusion alternative splicing. From this knowledge, we designed and tested a Droplet Digital PCR assay that detects CNTRL-FGFR1 expression to approximately one cell in 100 000 using fusion breakpoint-specific primers and probes. We also utilised cell-line models to show that effective tyrosine kinase inhibitors, which may be included in treatment regimens for this disease, are only those that block FGFR1 phosphorylation.
Collapse
Affiliation(s)
- Lauren M Brown
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Ray C Bartolo
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Nadia M Davidson
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia.,School of BioSciences, University of Melbourne, Parkville, Australia
| | - Breon Schmidt
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Ian Brooks
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Jackie Challis
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Vida Petrovic
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Dong-Anh Khuong-Quang
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Australia.,Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia
| | - Francoise Mechinaud
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia.,Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia
| | - Seong L Khaw
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia.,Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia.,Walter and Eliza Hall Institute, Parkville, Australia
| | - Ian J Majewski
- Walter and Eliza Hall Institute, Parkville, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Alicia Oshlack
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia.,School of BioSciences, University of Melbourne, Parkville, Australia
| | - Paul G Ekert
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Australia
| |
Collapse
|
19
|
Shomali W, Gotlib J. World Health Organization-defined eosinophilic disorders: 2019 update on diagnosis, risk stratification, and management. Am J Hematol 2019; 94:1149-1167. [PMID: 31423623 DOI: 10.1002/ajh.25617] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 12/16/2022]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of non-hematologic (secondary or reactive) and hematologic (primary, clonal) disorders with potential for end-organ damage. DIAGNOSIS Hypereosinophilia has generally been defined as a peripheral blood eosinophil count greater than 1.5 × 109 /L, and may be associated with tissue damage. After exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on a combination of various tests. They include morphologic review of the blood and marrow, standard cytogenetics, fluorescence in situ-hybridization, flow immunophenotyping, and T-cell clonality assessment to detect histopathologic or clonal evidence for an acute or chronic hematolymphoid neoplasm. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2016 World Health Organization endorses a semi-molecular classification scheme of disease subtypes. This includes the major category "myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB, or FGFR1 or with PCM1-JAK2", and the MPN subtype, "chronic eosinophilic leukemia, not otherwise specified" (CEL, NOS). Lymphocyte-variant hypereosinophilia is an aberrant T-cell clone-driven reactive eosinophila, and idiopathic hypereosinophilic syndrome (HES) is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (eg, <1.5 × 109 /L) without symptoms or signs of organ involvement, a watch and wait approach with close-follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Corticosteroids are first-line therapy for patients with lymphocyte-variant hypereosinophilia and HES. Hydroxyurea and interferon-alfa have demonstrated efficacy as initial treatment and in steroid-refractory cases of HES. In addition to hydroxyurea, second line cytotoxic chemotherapy agents, and hematopoietic stem cell transplantation have been used for aggressive forms of HES and CEL, with outcomes reported for limited numbers of patients. The use of antibodies against interleukin-5 (IL-5) (mepolizumab), the IL-5 receptor (benralizumab), as well as other targets on eosinophils remains an active area of investigation.
Collapse
Affiliation(s)
- William Shomali
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, California
| | - Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, California
| |
Collapse
|
20
|
Fibroblast Growth Factor Receptor, a Novel Receptor for Vegetative Insecticidal Protein Vip3Aa. Toxins (Basel) 2018; 10:toxins10120546. [PMID: 30567360 PMCID: PMC6315849 DOI: 10.3390/toxins10120546] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 11/17/2022] Open
Abstract
Vegetative insecticidal proteins (Vips), which are secreted by some Bacillus thuringiensis strains during vegetative growth, exhibit high virulence to many pests. Vip3A proteins have been used commercially both in some bio-insecticides and in transgenic crops; however, compared with insecticidal crystal proteins, the mechanism of action of Vip3A is still unclear. In this work, we indicated that the fibroblast growth factor receptor-like protein (Sf-FGFR) from the membrane of Sf9 cells could bind to Vip3Aa. The interaction between Vip3Aa and Sf-FGFR was confirmed by pull-down assays and dot blotting experiment in vitro. The binding affinity between Vip3Aa and extracellular regions of Sf-FGFR (GST-FGFR-N) was determined by microscale thermophoresis assay (MST). Moreover, Vip3Aa-Flag could be co-immunoprecipitated with Sf-FGFR-V5 ex vivo. Furthermore, knockdown of Sf-FGFR gene in Sf9 cells resulted in reducing the mortality of those cells to Vip3Aa. In summary, our data indicated that Sf-FGFR is a novel receptor for Vip3Aa.
Collapse
|
21
|
Zhang XW, Chen MY, Chao HY, Liu J, Chen SN, Zhang R, Zhou M, Lu XZ, Wang Q. [Clinical and molecular features of one case of 8p11 myeloproliferative syndrome with t(8;17) (p11; q24)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:1036-1039. [PMID: 30612409 PMCID: PMC7348216 DOI: 10.3760/cma.j.issn.0253-2727.2018.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Indexed: 11/29/2022]
Affiliation(s)
| | | | - H Y Chao
- Department of Hematology, Affiliated Changzhou Second Hospital of NanJing Medical University, Changzhou 213003, China
| | | | | | | | | | | | | |
Collapse
|
22
|
Cao M, Carrasco RD, Dubuc AM, Dal Cin P, Fletcher JA, Xiao S. ZMYM2-FGFR1 fusion as secondary change in acute myeloid leukemia. Leuk Lymphoma 2018; 60:556-558. [PMID: 30160587 DOI: 10.1080/10428194.2018.1493733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Minghua Cao
- a Department of Pathology, Brigham and Women's Hospital , Harvard Medical School , Boston ( MA) , USA
| | - Ruben D Carrasco
- a Department of Pathology, Brigham and Women's Hospital , Harvard Medical School , Boston ( MA) , USA
| | - Adrian M Dubuc
- a Department of Pathology, Brigham and Women's Hospital , Harvard Medical School , Boston ( MA) , USA
| | - Paola Dal Cin
- a Department of Pathology, Brigham and Women's Hospital , Harvard Medical School , Boston ( MA) , USA
| | - Jonathan A Fletcher
- a Department of Pathology, Brigham and Women's Hospital , Harvard Medical School , Boston ( MA) , USA
| | - Sheng Xiao
- a Department of Pathology, Brigham and Women's Hospital , Harvard Medical School , Boston ( MA) , USA
| |
Collapse
|
23
|
Myeloid and Lymphoid Neoplasms with Eosinophilia and Abnormalities of PDGFRA, PDGFRB, FGFR1, or t(8;9)(p22;p24.1);PCM1-JAK2. MOLECULAR PATHOLOGY LIBRARY 2018. [DOI: 10.1007/978-3-319-62146-3_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
24
|
Gotlib J. World Health Organization-defined eosinophilic disorders: 2017 update on diagnosis, risk stratification, and management. Am J Hematol 2017; 92:1243-1259. [PMID: 29044676 DOI: 10.1002/ajh.24880] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/03/2017] [Indexed: 12/20/2022]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of nonhematologic (secondary or reactive) and hematologic (primary, clonal) disorders with potential for end-organ damage. DIAGNOSIS Hypereosinophilia has generally been defined as a peripheral blood eosinophil count greater than 1500/mm3 and may be associated with tissue damage. After exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on a combination of morphologic review of the blood and marrow, standard cytogenetics, fluorescent in situ-hybridization, flow immunocytometry, and T-cell clonality assessment to detect histopathologic or clonal evidence for an acute or chronic myeloid or lymphoproliferative disorder. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2016 World Health Organization endorses a semi-molecular classification scheme of disease subtypes which includes the major category "myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB, or FGFR1 or with PCM1-JAK2," and the "MPN subtype, chronic eosinophilic leukemia, not otherwise specified" (CEL, NOS). Lymphocyte-variant hypereosinophilia is an aberrant T-cell clone-driven reactive eosinophila, and idiopathic hypereosinophilic syndrome (HES) is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (e.g., < 1500/mm3 ) without symptoms or signs of organ involvement, a watch and wait approach with close-follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Corticosteroids are first-line therapy for patients with lymphocyte-variant hypereosinophilia and HES. Hydroxyurea and interferon-alpha have demonstrated efficacy as initial treatment and steroid-refractory cases of HES. In addition to hydroxyurea, second line cytotoxic chemotherapy agents and hematopoietic cell transplant have been used for aggressive forms of HES and CEL with outcomes reported for limited numbers of patients. The use of antibodies against interleukin-5 (IL-5) (mepolizumab), the IL-5 receptor (benralizumab), and CD52 (alemtuzumab), as well as other targets on eosinophils remains an active area of investigation.
Collapse
Affiliation(s)
- Jason Gotlib
- Stanford Cancer Institute, Stanford, California 94305-5821
| |
Collapse
|
25
|
Gotlib J. Tyrosine Kinase Inhibitors in the Treatment of Eosinophilic Neoplasms and Systemic Mastocytosis. Hematol Oncol Clin North Am 2017; 31:643-661. [PMID: 28673393 DOI: 10.1016/j.hoc.2017.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The World Health Organization's semimolecular classification of eosinophilias emphasizes neoplasms driven by fusion tyrosine kinases. More than 80% of patients with systemic mastocytosis carry the KIT D816V mutation, the primary driver of disease pathogenesis. Genetic annotation of these diseases is critical and affords opportunities for targeted therapy. This article discusses our understanding of the mutated tyrosine kinome of eosinophilic neoplasms and systemic mast cell disease, and the successes and limitations of available therapies. Use of tyrosine kinase inhibitors as a bridge to hematopoietic stem cell transplantation, and development of more selective and potent tyrosine kinase inhibitors is also highlighted.
Collapse
Affiliation(s)
- Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, 875 Blake Wilbur Drive, Room 2324, Stanford, CA 94305-5821, USA.
| |
Collapse
|
26
|
Landberg N, Dreimane A, Rissler M, Billström R, Ågerstam H. Primary cells inBCR/FGFR1-positive 8p11 myeloproliferative syndrome are sensitive to dovitinib, ponatinib, and dasatinib. Eur J Haematol 2017; 99:442-448. [DOI: 10.1111/ejh.12957] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Niklas Landberg
- Department of Clinical Genetics; Lund University; Lund Sweden
| | - Arta Dreimane
- Department of Haematology; Linköping University Hospital; Linköping Sweden
| | | | - Rolf Billström
- Department of Medicine; Central Hospital Skövde; Skövde Sweden
| | - Helena Ågerstam
- Department of Clinical Genetics; Lund University; Lund Sweden
| |
Collapse
|
27
|
Cowell JK, Qin H, Hu T, Wu Q, Bhole A, Ren M. Mutation in the FGFR1 tyrosine kinase domain or inactivation of PTEN is associated with acquired resistance to FGFR inhibitors in FGFR1-driven leukemia/lymphomas. Int J Cancer 2017. [PMID: 28646488 DOI: 10.1002/ijc.30848] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Stem cell leukemia/lymphoma syndrome (SCLL) is driven by constitutive activation of chimeric FGFR1 kinases generated by chromosome translocations. We have shown that FGFR inhibitors significantly suppress leukemia and lymphoma development in vivo, and cell viability in vitro. Since resistance to targeted therapies is a major reason for relapse, we developed FGFR1-overexpressing mouse and human cell lines that are resistant to the specific FGFR inhibitors AZD4547 and BGJ398, as well as non-specific inhibitors, such as ponatinib, TKI258 and E3810. Two mutually exclusive mechanisms for resistance were demonstrated; an activating V561M mutation in the FGFR1 kinase domain and mutational inactivation of PTEN resulting in increased PI3K/AKT activity. Ectopic expression of PTEN in the PTEN-mutant cells resensitizes them to FGFR inhibitors. Treatment of resistant cells with BGJ398, in combination with the BEZ235 PI3K inhibitor, shows an additive effect on growth in vitro and prolongs survival in xenograft models in vivo. These studies provide the first direct evidence for both the involvement of the FGFR1 V561M mutation and PTEN inactivation in the development of resistance in leukemias overexpressing chimeric FGFR1. These studies also provide a potential strategy to treat leukemias and lymphomas driven by FGFR1 activation that become resistant to FGFR1 inhibitors.
Collapse
Affiliation(s)
- John K Cowell
- Georgia Cancer Center, Augusta University, Augusta, GA
| | - Haiyan Qin
- Georgia Cancer Center, Augusta University, Augusta, GA
| | - Tianxiang Hu
- Georgia Cancer Center, Augusta University, Augusta, GA
| | - Qing Wu
- Georgia Cancer Center, Augusta University, Augusta, GA
| | - Aaron Bhole
- Georgia Cancer Center, Augusta University, Augusta, GA
| | - Mingqiang Ren
- Georgia Cancer Center, Augusta University, Augusta, GA
| |
Collapse
|
28
|
Sarthy JF, Reddivalla N, Radhi M, Chastain K. Pediatric 8p11 eosinophilic myeloproliferative syndrome (EMS): A case report and review of the literature. Pediatr Blood Cancer 2017; 64. [PMID: 27808462 DOI: 10.1002/pbc.26310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/05/2016] [Accepted: 09/21/2016] [Indexed: 01/05/2023]
Abstract
The 8p11 eosinophilic myeloproliferative syndrome (EMS) is an aggressive neoplasm driven by translocation of the fibroblast growth factor receptor 1 and often transforms to leukemias and lymphomas that are refractory to treatment. The first case was identified in 1983, and to date over 70 cases have been reported in the literature. Despite those reports, no consensus exists on management of this condition, and inconsistency in treatment regimens is even more pronounced in the pediatric literature. We report a case of a male infant with the 8p11 EMS, review the published pediatric experience with EMS, and discuss treatment strategies for this enigmatic hematological disorder.
Collapse
Affiliation(s)
- Jay F Sarthy
- Children's Mercy Hospitals and Clinics, Kansas City, Missouri 64113
| | | | - Mohamed Radhi
- Children's Mercy Hospitals and Clinics, Kansas City, Missouri 64113
| | | |
Collapse
|
29
|
Abstract
Abstract
Molecular diagnostics has generated substantial dividends in dissecting the genetic basis of myeloid neoplasms with eosinophilia. The family of diseases generated by dysregulated fusion tyrosine kinase (TK) genes is recognized by the World Health Organization (WHO) category, “Myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB, or FGFR1, or with PCM1-JAK2.” In addition to myeloproliferative neoplasms (MPN), these patients can present with myelodysplastic syndrome/MPN, as well as de novo or secondary mixed-phenotype leukemias or lymphomas. Eosinophilia is a common, but not invariable, feature of these diseases. The natural history of PDGFRA- and PDGFRB-rearranged neoplasms has been dramatically altered by imatinib. In contrast, patients with FGFR1 and JAK2 fusion TK genes exhibit a more aggressive course and variable sensitivity to current TK inhibitors, and in most cases, long-term disease-free survival may only be achievable with allogeneic hematopoietic stem cell transplantation. Similar poor prognosis outcomes may be observed with rearrangements of FLT3 or ABL1 (eg, both of which commonly partner with ETV6), and further investigation is needed to validate their inclusion in the current WHO-defined group of eosinophilia-associated TK fusion-driven neoplasms. The diagnosis chronic eosinophilic leukemia, not otherwise specified (CEL, NOS) is assigned to patients with MPN with eosinophilia and nonspecific cytogenetic/molecular abnormalities and/or increased myeloblasts. Myeloid mutation panels have identified somatic variants in patients with a provisional diagnosis of hypereosinophilia of undetermined significance, reclassifying some of these cases as eosinophilia-associated neoplasms. Looking forward, one of the many challenges will be how to use the results of molecular profiling to guide prognosis and selection of actionable therapeutic targets.
Collapse
|
30
|
Xie X, Wang Z, Chen F, Yuan Y, Wang J, Liu R, Chen Q. Roles of FGFR in oral carcinogenesis. Cell Prolif 2017; 49:261-9. [PMID: 27218663 DOI: 10.1111/cpr.12260] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/29/2016] [Indexed: 12/12/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) play essential roles in organ development during the embryonic period, and regulate tissue repair in adults. Accumulating evidence suggests that alterations in FGFR signalling are involved in diverse types of cancer. In this review, we focus on aberrant regulation of FGFRs in pathogenesis of oral squamous cell carcinoma (OSCC), including altered expression and subcellular location, aberrant isoform splicing and mutations. We also provide an overview of oncogenic roles of each FGFR and its downstream signalling pathways in regulating OSCC cell proliferation and metastasis. Finally, we discuss potential application of FGFRs as anti-cancer targets in the preclinical environment and in clinical practice.
Collapse
Affiliation(s)
- Xiaoyan Xie
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Fangman Chen
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yao Yuan
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiayi Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
31
|
Qiu XH, Li F, Cao HQ, Shao JJ, Mei JG, Li HQ, Zhai YP. Activity of fibroblast growth factor receptor inhibitors TKI258, ponatinib and AZD4547 against TPR‑FGFR1 fusion. Mol Med Rep 2017; 15:1024-1030. [PMID: 28138694 PMCID: PMC5367333 DOI: 10.3892/mmr.2017.6140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 11/29/2016] [Indexed: 12/21/2022] Open
Abstract
8p11 myeloproliferative syndrome (EMS) is a rare disease characterized by the constitutive activation of fibroblast growth factor receptor 1 (FGFR1). To date, four cases of EMS with the chromosomal translocation, t(1;8)(q25;p11.2), have been reported. In the present study, TPR-FGFR1-expressing Baf3 cells were established and confirmed by polymerase chain reaction. To identify the most promising drug for EMS, the activities and associated mechanism of three tyrosine kinase inhibitors (TKIs), TKI258, ponatinib and AZD4547, against TPR-FGFR1 were tested by MTT assay, flow cytometry and western blot. The data demonstrated that TPR-FGFR1 was localized in the cytoplasm, and was able to transform interleukin-3-dependent hematopoietic Baf3 cells into growth factor-independent cells. All of the three TKIs markedly inhibited the proliferation of TPR-FGFR1-expressing Baf3 cells, and the activation of FGFR1 and the downstream signaling molecules, extracellular signal-regulated kinase 1/2, phospholipiase Cγ and signal transducer and activator of transcription 5. AZD4547 was the most efficient drug, and TKI258 was the least. By contrast, no significant difference was found among the three drugs on their effect on cell apoptosis. Taken together, the data obtained in the present study suggested that AZD4547 had increased potency, compared with TKI258 and ponatinib, for the treatment of EMS.
Collapse
Affiliation(s)
- Xu-Hua Qiu
- Department of Hematology, Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Feng Li
- Department of Hematology, Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Hong-Qin Cao
- Department of Hematology, Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Jing-Jing Shao
- Department of Hematology, Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Jian-Gang Mei
- Department of Hematology, Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Han-Qing Li
- Department of Hematology, Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Yong-Ping Zhai
- Department of Hematology, Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
32
|
Nelson KN, Peiris MN, Meyer AN, Siari A, Donoghue DJ. Receptor Tyrosine Kinases: Translocation Partners in Hematopoietic Disorders. Trends Mol Med 2016; 23:59-79. [PMID: 27988109 DOI: 10.1016/j.molmed.2016.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/11/2016] [Accepted: 11/13/2016] [Indexed: 02/07/2023]
Abstract
Receptor tyrosine kinases (RTKs) activate various signaling pathways and regulate cellular proliferation, survival, migration, and angiogenesis. Malignant neoplasms often circumvent or subjugate these pathways by promoting RTK overactivation through mutation or chromosomal translocation. RTK translocations create a fusion protein containing a dimerizing partner fused to an RTK kinase domain, resulting in constitutive kinase domain activation, altered RTK cellular localization, upregulation of downstream signaling, and novel pathway activation. While RTK translocations in hematological malignancies are relatively rare, clinical evidence suggests that patients with these genetic abnormalities benefit from RTK-targeted inhibitors. Here, we present a timely review of an exciting field by examining RTK chromosomal translocations in hematological cancers, such as Anaplastic Lymphoma Kinase (ALK), Fibroblast Growth Factor Receptor (FGFR), Platelet-Derived Growth Factor Receptor (PDGFR), REarranged during Transfection (RET), Colony Stimulating Factor 1 Receptor (CSF1R), and Neurotrophic Tyrosine Kinase Receptor Type 3 (NTRK3) fusions, and discuss current therapeutic options.
Collapse
Affiliation(s)
- Katelyn N Nelson
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Malalage N Peiris
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - April N Meyer
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Asma Siari
- Université Joseph Fourier Grenoble, Grenoble, France
| | - Daniel J Donoghue
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA; Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
33
|
Tsyba NN, Turkina AG, Chelysheva EY, Nemchenko IS, Kovrigina AM, Obukhova TN, Urnova ES, Kuzmina LA, Savchenko VG. [A rare case of myeloproliferative disease with t(8;13)(p11;q12) associated with eosinophilia and lymphadenopathy]. TERAPEVT ARKH 2016; 88:98-103. [PMID: 27459622 DOI: 10.17116/terarkh201688798-103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Myeloproliferative disease associated with FGFR1 rearrangement (8p11), which is included in the 2008 WHO Classification of Myeloid Neoplasms, is a rare and extremely aggressive abnormality. The paper describes a clinical case of a 39-year-old female patient who was detected to have leukocytosis (as high as 47.2·109/l), absolute eosinophilia (as high as 3.1·109/l), and enlarged peripheral lymph nodes during her visit to a doctor. The bone marrow (BM) showed the changes typically encountered in myeloproliferative disease with eosinophilia. The patient was found to have t(8;13)(p11;q12) translocation associated with the rearrangement of the FGFR1 gene located at the 8p11 locus. Molecular and cytogenetic examinations failed to reveal BCR-ABL chimeric transcript, Jak2 V617F mutation, and deletions and translocations involving PDGFRA (4q12) and PDGFRB (5q32-33). The similar changes in the karyotype were also found in the lymph node cells. The undertaken treatment with hydroxyurea and the tyrosine kinase inhibitor dasatinib turned out to be ineffective. The patient underwent allogeneic BM transplantation from a HLA-identical sibling. Graft rejection occurred 6 months later. Allogeneic BM transplantation from the same donor (100% donor chimerism; FGFR1/8р11 translocation was not detected), which was complicated by the development of chronic graft-versus-host reaction, was performed again in March 2015. The patient is being followed up and continues to receive immunosuppressive therapy.
Collapse
Affiliation(s)
- N N Tsyba
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - A G Turkina
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - E Yu Chelysheva
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - I S Nemchenko
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - A M Kovrigina
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - T N Obukhova
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - E S Urnova
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - L A Kuzmina
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| | - V G Savchenko
- National Research Center for Hematology, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
34
|
Ren M, Qin H, Wu Q, Savage NM, George TI, Cowell JK. Development of ZMYM2-FGFR1 driven AML in human CD34+ cells in immunocompromised mice. Int J Cancer 2016; 139:836-40. [PMID: 27005999 DOI: 10.1002/ijc.30100] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/22/2016] [Accepted: 03/17/2016] [Indexed: 12/14/2022]
Abstract
Acute myelogenous leukemia (AML) has an overall poor survival rate and shows considerable molecular heterogeneity in its etiology. In the WHO classification there are >50 cytogenetic subgroups of AML, many showing highly specific chromosome translocations that lead to constitutive activation of individual kinases. In a rare stem cell leukemia/lymphoma syndrome, translocations involving 8p11 lead to constitutive activation of the fibroblast growth factor receptor 1 (FGFR1) kinase. This disorder shows myeloproliferative disease with almost invariable progresses to AML and conventional therapeutic strategies are largely unsuccessful. Because of the rare nature of this syndrome, models that faithfully recapitulate the human disease are needed to evaluate therapeutic strategies. The t(8;13)(p11;q12) chromosome translocation is most common rearrangement seen in this syndrome and creates a ZMYM2-FGFR1 chimeric kinase. To understand more about the molecular etiology of AML induced by this particular rearrangement, we have created a model human CD34+ cells transplanted into immunocompromized mice which develop myeloproliferative disease that progresses to AML with a long (>12 months) latency period. As in humans, these mice show hepatospenomegaly, hypercellular bone marrow and a CD45 + CD34 + CD13+ immunophenotype. Molecular studies demonstrate upregulation of genes such as KLF4 and FLT3 that promote stemness, and overexpression of MYC, which is associated with suppression of myeloid cell differentiation. This murine model, therefore, provides an opportunity to develop therapeutic strategies against the most common subtype within these FGFR1 driven neoplasms and study the molecular etiology in more depth.
Collapse
Affiliation(s)
- Mingqiang Ren
- Georgia Regents University Cancer Center, Augusta, GA
| | - Haiyan Qin
- Georgia Regents University Cancer Center, Augusta, GA
| | - Qing Wu
- Georgia Regents University Cancer Center, Augusta, GA
| | - Natasha M Savage
- Georgia Regents University Cancer Center, Augusta, GA.,GRU Department of Pathology, Augusta, GA
| | - Tracy I George
- Department of Pathology, University of New Mexico, Albuquerque, NM
| | - John K Cowell
- Georgia Regents University Cancer Center, Augusta, GA
| |
Collapse
|
35
|
Abstract
Eosinophilia in the peripheral blood can be the manifestation various medical conditions, including benign or malignant disorders. There are 3 main types of eosinophilia-associated myeloid neoplasms (MN-eos): myeloid and lymphoid neoplasms, chronic eosinophilic leukemia not otherwise specified, and idiopathic hypereosinophilic syndrome (HES). Imatinib mesylate has revolutionized the treatment of molecularly defined MN-eos, and novel agents have been successfully used to treat HES. The discovery of new, recurrent molecular alterations in patients with MN-eos may improve their diagnosis and therapy. This review focuses on the hematologist's approach to a patient with eosinophilia and treatment options for those with eosinophilic myeloid neoplasms.
Collapse
Affiliation(s)
- Lorenzo Falchi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
36
|
Gotlib J. World Health Organization-defined eosinophilic disorders: 2015 update on diagnosis, risk stratification, and management. Am J Hematol 2015; 90:1077-89. [PMID: 26486351 DOI: 10.1002/ajh.24196] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 09/15/2015] [Indexed: 12/19/2022]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of non-hematologic (secondary or reactive) and hematologic (primary, clonal) disorders with potential for end-organ damage. DIAGNOSIS Hypereosinophilia has generally been defined as a peripheral blood eosinophil count greater than 1,500/mm(3) and may be associated with tissue damage. After exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on a combination of morphologic review of the blood and marrow, standard cytogenetics, fluorescent in situ-hybridization, flow immunocytometry, and T-cell clonality assessment to detect histopathologic or clonal evidence for an acute or chronic myeloid or lymphoproliferative disorder. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2008 World Health Organization establishes a semi-molecular classification scheme of disease subtypes including 'myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB, or FGFR1', chronic eosinophilic leukemia, not otherwise specified, (CEL, NOS), lymphocyte-variant hypereosinophilia, and idiopathic hypereosinophilic syndrome (HES), which is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of the therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (e.g. < 1,500/mm(3) ) without symptoms or signs of organ involvement, a watch and wait approach with close-follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Corticosteroids are first-line therapy for patients with lymphocyte-variant hypereosinophilia and HES. Hydroxyurea and interferon-alpha have demonstrated efficacy as initial treatment and steroid-refractory cases of HES. In addition to hydroxyurea, second line cytotoxic chemotherapy agents and hematopoietic cell transplant have been used for aggressive forms of HES and CEL with outcomes reported for limited numbers of patients. Although clinical trials have been performed with anti IL-5 (mepolizumab) and anti-CD52 (alemtuzumab) antibodies, their therapeutic role in primary eosinophilic diseases and HES has yet to be established.
Collapse
|
37
|
Zhu N, Xiao H, Wang LM, Fu S, Zhao C, Huang H. Mutations in tyrosine kinase and tyrosine phosphatase and their relevance to the target therapy in hematologic malignancies. Future Oncol 2015; 11:659-73. [PMID: 25686120 DOI: 10.2217/fon.14.280] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Protein tyrosine kinases and protein tyrosine phosphatases play pivotal roles in regulation of cellular phosphorylation and signal transduction with opposite functions. Accumulating evidences have uncovered the relevance of genetic alterations in these two family members to hematologic malignancies. This review underlines progress in understanding the pathogenesis of these genetic alterations including mutations and aberrant expression and the evolving protein tyrosine kinases and protein tyrosine phosphatases targeted therapeutic strategies in hematologic neoplasms.
Collapse
Affiliation(s)
- Ni Zhu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China
| | | | | | | | | | | |
Collapse
|
38
|
Gotlib J. Tyrosine Kinase Inhibitors and Therapeutic Antibodies in Advanced Eosinophilic Disorders and Systemic Mastocytosis. Curr Hematol Malig Rep 2015; 10:351-61. [DOI: 10.1007/s11899-015-0280-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
39
|
Clinical activity of ponatinib in a patient with FGFR1-rearranged mixed-phenotype acute leukemia. Leukemia 2015; 30:947-50. [PMID: 26055304 DOI: 10.1038/leu.2015.136] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
40
|
Kumar KR, Chen W, Koduru PR, Luu HS. Myeloid and lymphoid neoplasm with abnormalities of FGFR1 presenting with trilineage blasts and RUNX1 rearrangement: a case report and review of literature. Am J Clin Pathol 2015; 143:738-48. [PMID: 25873510 DOI: 10.1309/ajcpud6w1jlqqmna] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Myeloid and lymphoid neoplasms with abnormalities of fibroblast growth factor receptor 1 gene (FGFR1) are a rare and aggressive disease group that harbors translocations of FGFR1 with at least 14 recognized partner genes. We report a case of a patient with a novel t(17;21)(p13;q22) with RUNX1 rearrangement and trilineage blasts. METHODS A 29-year-old man with relapsed T-lymphoblastic lymphoma in the cervical nodes showed a myeloproliferative neoplasm in his bone marrow with three separate populations of immunophenotypically aberrant myeloid, T-lymphoid, and B-lymphoid blasts by flow cytometry. Cytogenetic and fluorescent in situ hybridization studies showed unique dual translocations of t(8;13)(p11.2;q12) and t(17;21)(p13;q22) with RUNX1 rearrangement. RESULTS The patient was initiated on a mitoxantrone, etoposide, and cytarabine chemotherapy regimen and died of complications of disease 1 month later. CONCLUSIONS To our knowledge, this is the first reported case of a myeloid and lymphoid neoplasm with abnormalities of FGFR1 with t(17;21)(p13;q22) and trilineage blasts.
Collapse
Affiliation(s)
- Kirthi R. Kumar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas
| | - Weina Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas
| | - Prasad R. Koduru
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas
| | - Hung S. Luu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
41
|
Kiflemariam S, Ljungström V, Pontén F, Sjöblom T. Tumor vessel up-regulation of INSR revealed by single-cell expression analysis of the tyrosine kinome and phosphatome in human cancers. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1600-9. [PMID: 25864925 DOI: 10.1016/j.ajpath.2015.02.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 02/08/2015] [Accepted: 02/18/2015] [Indexed: 01/16/2023]
Abstract
The tyrosine kinome and phosphatome harbor oncogenes and tumor suppressor genes and important regulators of angiogenesis and tumor stroma formation. To provide a better understanding of their potential roles in cancer, we analyzed the expression of 85 tyrosine kinases and 42 tyrosine phosphatases by in situ hybridization 48 human normal and 24 tumor tissue specimens. Nine-tenths of the assessed transcripts had tumor cell expression concordant with expression array databases. Further, pan-cancer expression of AATK, PTPRK, and PTPRU and expression of PTPRS in a subset of tumors were observed. To demonstrate tumor subcompartment resolution, we validated the predicted tumor stroma-specific markers HTRA1, HTRA3, MXRA5, MXRA8, and SERPING1 in situ. In addition to known vascular and stromal markers such as PDGFRB, we observed stromal expression of PTK6 and TNS1 and vascular expression of INSR, PTPRF, PTPRG, PTPRU, and TNS1, of which INSR emerged as a tumor-specific vessel marker. This study demonstrates the feasibility of large-scale analyses to chart the transcriptome in situ in human cancers and their ability to identify novel cancer biomarkers.
Collapse
Affiliation(s)
- Sara Kiflemariam
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Viktor Ljungström
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fredrik Pontén
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Tobias Sjöblom
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
42
|
Zhou F, Chen S, Chao H, Zhang R, Zhou M, Pan J. [Clinical and gene involved of one case of 8p11 myeloproliferative syndrome with ins(13;8)(q12;p11p23)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:291-6. [PMID: 25916288 PMCID: PMC7342628 DOI: 10.3760/cma.j.issn.0253-2727.2015.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To improve the understanding of patients with 8p11 myeloproliferative syndrome (EMS) harboring ins(13;8)(q12;p11p23)/ZNF198 -FGFR1. METHODS We reported here a 8p11 EMS case and provided more details on the clinical and molecular features of ins(13;8)(q12;p11p23)/ZNF198-FGFR1,full length ZNF198-FGFR1 was cloned by overlap extension PCR method,and the literatures on this topic were reviewed. RESULTS Clinically, the case with ins(13;8)(q12;p11p23)/ZNF198-FGFR1 had distinct hematological and clinical characteristics: hyperleukocytosis, myeloid hyperplasia,widespread adenopathy and lymphoma; Fluorescence in situ hybridization (FISH) disclosed the positive FGFR1 gene rearrangement; Further molecular studies confirmed a mRNA in-frame fusion between exon 17 of the ZNF198 gene and exon 9 of FGFR1 gene ,the full length ZNF198-FGFR1 was composed of a NH2 terminus of ZNF198 including the ZNF and proline-rich domains, whereas the COOH terminus of FGFR1 included 2 tyrosine kinase domains. CONCLUSION EMS with ins(13;8)(q12;p11p23)/ZNF198 -FGFR1 was a very rare, distinct myeloproliferative neoplasm, the fusion gene and chimeric protein with constitutive activation of the FGFR1 tyrosine kinase.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Hematology, Affiliated Changzhou Second Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Suning Chen
- Department of Hematology, Affiliated Changzhou Second Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Hongying Chao
- Department of Hematology, Affiliated Changzhou Second Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Ri Zhang
- Department of Hematology, Affiliated Changzhou Second Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Min Zhou
- Department of Hematology, Affiliated Changzhou Second Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Jinlan Pan
- Department of Hematology, Affiliated Changzhou Second Hospital of Nanjing Medical University, Changzhou 213003, China
| |
Collapse
|
43
|
Yamamoto S, Otsu M, Matsuzaka E, Konishi C, Takagi H, Hanada S, Mochizuki S, Nakauchi H, Imai K, Tsuji K, Ebihara Y. Screening of drugs to treat 8p11 myeloproliferative syndrome using patient-derived induced pluripotent stem cells with fusion gene CEP110-FGFR1. PLoS One 2015; 10:e0120841. [PMID: 25803811 PMCID: PMC4372437 DOI: 10.1371/journal.pone.0120841] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 02/10/2015] [Indexed: 01/01/2023] Open
Abstract
Induced pluripotent stem (iPS) cells provide powerful tools for studying disease mechanisms and developing therapies for diseases. The 8p11 myeloproliferative syndrome (EMS) is an aggressive chronic myeloproliferative disorder (MPD) that is caused by constitutive activation of fibroblast growth factor receptor 1. EMS is rare and, consequently, effective treatment for this disease has not been established. Here, iPS cells were generated from an EMS patient (EMS-iPS cells) to assist the development of effective therapies for EMS. When iPS cells were co-cultured with murine embryonic stromal cells, EMS-iPS cells produced more hematopoietic progenitor and hematopoietic cells, and CD34+ cells derived from EMS-iPS cells exhibited 3.2–7.2-fold more macrophage and erythroid colony forming units (CFUs) than those derived from control iPS cells. These data indicate that EMS-iPS cells have an increased hematopoietic differentiation capacity, which is characteristic of MPDs. To determine whether a tyrosine kinase inhibitor (TKI) could suppress the increased number of CFUs formed by EMS-iPS-induced CD34+ cells, cells were treated with one of four TKIs (CHIR258, PKC 412, ponatinib, and imatinib). CHIR258, PKC 412, and ponatinib reduced the number of CFUs formed by EMS-iPS-induced CD34+ cells in a dose-dependent manner, whereas imatinib did not. Similar effects were observed on primary peripheral blood cells (more than 90% of which were blasts) isolated from the patient. This study provides evidence that the EMS-iPS cell line is a useful tool for the screening of drugs to treat EMS and to investigate the mechanism underlying this disease.
Collapse
Affiliation(s)
- Shohei Yamamoto
- Department of Pediatric Hematology/Oncology, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Stem Cell Processing, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Makoto Otsu
- Department of Pediatric Hematology/Oncology, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Stem Cell Processing, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Stem Cell Bank, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Emiko Matsuzaka
- Division of Stem Cell Processing, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Chieko Konishi
- Stem Cell Bank, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Haruna Takagi
- Stem Cell Bank, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sachiyo Hanada
- Division of Stem Cell Processing, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shinji Mochizuki
- Department of Pediatric Hematology/Oncology, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Stem Cell Processing, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kohzoh Imai
- Center for Antibody and Vaccine, IMSUT Hospital, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kohichiro Tsuji
- Department of Pediatric Hematology/Oncology, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- National Hospital Organization Shinshu Ueda Medical Center, Ueda, Japan
| | - Yasuhiro Ebihara
- Department of Pediatric Hematology/Oncology, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Stem Cell Processing, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Advanced Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
44
|
Chao H, Zhou M, Zhang R. A case with ZNF198-FGFR1 gene rearrangement presenting as acute eosinophil myeloid leukemia. Chin Med J (Engl) 2015; 128:131-2. [PMID: 25563327 PMCID: PMC4837809 DOI: 10.4103/0366-6999.147863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
| | | | - Ri Zhang
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| |
Collapse
|
45
|
Radonjic-Hoesli S, Valent P, Klion AD, Wechsler ME, Simon HU. Novel targeted therapies for eosinophil-associated diseases and allergy. Annu Rev Pharmacol Toxicol 2014; 55:633-56. [PMID: 25340931 DOI: 10.1146/annurev-pharmtox-010814-124407] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Eosinophil-associated diseases often present with life-threatening manifestations and/or chronic organ damage. Currently available therapeutic options are limited to a few drugs that often have to be prescribed on a lifelong basis to keep eosinophil counts under control. In the past 10 years, treatment options and outcomes in patients with clonal eosinophilic and other eosinophilic disorders have improved substantially. Several new targeted therapies have emerged, addressing different aspects of eosinophil expansion and inflammation. In this review, we discuss available and currently tested agents as well as new strategies and drug targets relevant to both primary and secondary eosinophilic diseases, including allergic disorders.
Collapse
|
46
|
Kim WS, Park SG, Park G, Jang SJ, Moon DS, Kang SH. 8p11 myeloproliferative syndrome with t(1;8)(q25;p11.2): a case report and review of the literature. Acta Haematol 2014; 133:101-5. [PMID: 25227135 DOI: 10.1159/000363441] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/06/2014] [Indexed: 11/19/2022]
Abstract
8p11 myeloproliferative syndrome (EMS) is a rare disease characterized by myeloproliferative neoplasm (MPN) associated with eosinophilia and T or B lymphoblastic lymphoma/leukemia. EMS is defined by molecular disruption of the FGFR1 gene at the 8p11-12 chromosome locus, and various partner genes are associated with FGFR1 gene translocation or insertion. The different partner-FGFR1 fusion genes are associated with slightly different disease phenotypes. The present patient showed T lymphoblastic lymphoma in a cervical lymph node, involvement of malignant lymphoma in the skin, and MPN bone marrow morphology with peripheral monocytosis. Chromosome analysis of the patient showed t(1;8)(q25;p11.2). To our knowledge, only 2 cases of EMS with translocation of t(1;8)(q25;p11.2) have been previously reported. Including this case, all 3 cases with EMS with t(1;8)(q25;p11.2) showed MPN bone marrow morphology and peripheral monocytosis. These findings support that t(1;8)(q25;p11.2) is associated with peripheral monocytosis in EMS patients. Of the 2 cases of EMS with t(1;8)(q25;p11.2) which were previously reported, FGFR1 rearrangement was not confirmed in 1 case. Similarly, FGFR1 rearrangement in the present case was not detected by fluorescence in situ hybridization or reverse transcription-polymerase chain reaction. Further study is needed to identify other techniques that could be used to demonstrate FGFR1 rearrangement.
Collapse
Affiliation(s)
- Woo-Seong Kim
- Department of Laboratory Medicine, Chosun University College of Medicine, Gwangju, South Korea
| | | | | | | | | | | |
Collapse
|
47
|
Bedussi F, Bottini A, Memo M, Fox SB, Sigala S, Generali D. Targeting fibroblast growth factor receptor in breast cancer: a promise or a pitfall? Expert Opin Ther Targets 2014; 18:665-78. [PMID: 24833241 DOI: 10.1517/14728222.2014.898064] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Fibroblast growth factors (FGFs) along with their receptors (FGFRs) are involved in several cellular functions, from embryogenesis to metabolism. Because of the ability of FGFR signalling to induce cell proliferation, migration and survival in cancer, these have been found to become overactivated by several mechanisms, including gene amplification, chromosomal translocation and mutations. New evidences indicate that FGFs and FGFRs may act in an oncogenic fashion to promote multiple steps of cancer progression by inducing mitogenic and survival signals, as well as promoting epithelial-to-mesenchymal transition, invasion and tumour angiogenesis. This review focuses on the predictive and prognostic role of FGFRs, the role of FGFR signalling and how it may be most appropriately therapeutically targeted in breast cancer. AREAS COVERED Activation of the FGFR pathway is a common event in many cancer types and for this reason FGFR is an important potential target in cancer treatment. Relevant literature was reviewed to identify current and future role of FGFR family as a possible guide for selecting those patients who would be poor or good responders to the available or the upcoming target therapies for breast cancer treatment. EXPERT OPINION The success of a personalised medicine approach using targeted therapies ultimately depends on being capable of identifying the patients who will benefit the most from any given drug. Outlining the molecular mechanisms of FGFR signalling and discussing the role of this pathway in breast cancer, we would like to endorse the incorporation of specific patient selection biomakers with the rationale for therapeutic intervention with FGFR-targeted therapy in breast cancer.
Collapse
Affiliation(s)
- Francesca Bedussi
- University of Brescia Medical School, Department of Molecular and Translational Medicine, Section of Pharmacology , Brescia , Italy
| | | | | | | | | | | |
Collapse
|
48
|
Gotlib J. World Health Organization-defined eosinophilic disorders: 2014 update on diagnosis, risk stratification, and management. Am J Hematol 2014; 89:325-37. [PMID: 24577808 DOI: 10.1002/ajh.23664] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/03/2014] [Indexed: 12/27/2022]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of nonhematologic (secondary or reactive) and hematologic (primary, clonal) disorders with potential for end-organ damage. DIAGNOSIS Hypereosinophilia (HE) has generally been defined as a peripheral blood eosinophil count greater than 1,500/mm(3) and may be associated with tissue damage. After exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on a combination of morphologic review of the blood and marrow, standard cytogenetics, fluorescent in situ hybridization, flow immunocytometry, and T-cell clonality assessment to detect histopathologic or clonal evidence for an acute or chronic myeloid or lymphoproliferative disorder. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2008 World Health Organization establishes a semimolecular classification scheme of disease subtypes including "myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB, or FGFR1', chronic eosinophilic leukemia, not otherwise specified" (CEL, NOS), lymphocyte-variant HE, and idiopathic hypereosinophilic syndrome (HES), which is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (e.g., <1,500/mm(3)) without symptoms or signs of organ involvement, a watch and wait approach with close-follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Corticosteroids are first-line therapy for patients with lymphocyte-variant HE and HES. Hydroxyurea and interferon-alpha have demonstrated efficacy as initial treatment and steroid-refractory cases of HES. In addition to hydroxyurea, second-line cytotoxic chemotherapy agents and hematopoietic cell transplant have been used for aggressive forms of HES and CEL with outcomes reported for limited number of patients. Although clinical trials have been performed with anti-IL-5 (mepolizumab) and anti-CD52 (alemtuzumab) antibodies, their therapeutic role in primary eosinophilic diseases and HES has yet to be established.
Collapse
Affiliation(s)
- Jason Gotlib
- Division of Hematology; Stanford Cancer Center; Stanford California
| |
Collapse
|
49
|
Duckworth CB, Zhang L, Li S. Systemic mastocytosis with associated myeloproliferative neoplasm with t(8;19)(p12;q13.1) and abnormality of FGFR1: report of a unique case. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:801-807. [PMID: 24551307 PMCID: PMC3925931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 12/23/2013] [Indexed: 06/03/2023]
Abstract
Systemic mastocytosis is a neoplastic proliferation of mast cells that frequently presents with associated clonal hematological non-mast cell lineage disease. Myeloid and lymphoid neoplasms with abnormalities of the FGFR1 gene are a heterogenous group of rare and aggressive hematopoietic stem cell disorders. About a dozen of chromosome changes involving the FGFR1 gene, presenting as myeloid or lymphoid neoplasms, have been described in the literature. To date, only 2 cases of myeloid and lymphoid neoplasms with abnormalities of the FGFR1 gene have been reported in association with systemic mastocytosis, one with t(8;13) and one with t(8;17) involving the FGFR1 gene. Here we describe another case of myeloproliferative neoplasm with chromosome translocation t(8;19) involving FGFR1 gene associated with systemic mastocytosis.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biopsy
- Bone Marrow Examination
- Chromosomes, Human, Pair 19
- Chromosomes, Human, Pair 8
- Gene Rearrangement
- Genetic Predisposition to Disease
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Karyotyping
- Male
- Mastocytosis, Systemic/genetics
- Mastocytosis, Systemic/metabolism
- Mastocytosis, Systemic/pathology
- Mastocytosis, Systemic/surgery
- Myeloproliferative Disorders/genetics
- Myeloproliferative Disorders/metabolism
- Myeloproliferative Disorders/pathology
- Myeloproliferative Disorders/surgery
- Phenotype
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Stem Cell Transplantation
- Translocation, Genetic
- Treatment Outcome
Collapse
Affiliation(s)
- Christina B Duckworth
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine Atlanta, GA, USA
| | - Linsheng Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine Atlanta, GA, USA
| | - Shiyong Li
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine Atlanta, GA, USA
| |
Collapse
|
50
|
EMS: the 8p11 myeloproliferative syndrome. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|