1
|
Lahera A, Vela-Martín L, Fernández-Navarro P, Llamas P, López-Lorenzo JL, Cornago J, Santos J, Fernández-Piqueras J, Villa-Morales M. PIM1 is a potential therapeutic target for the leukemogenic effects mediated by JAK/STAT pathway mutations in T-ALL/LBL. NPJ Precis Oncol 2024; 8:152. [PMID: 39033228 PMCID: PMC11271448 DOI: 10.1038/s41698-024-00638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 07/09/2024] [Indexed: 07/23/2024] Open
Abstract
Precursor T-cell neoplasms (T-ALL/LBL) are aggressive hematological malignancies that arise from the malignant transformation of immature thymocytes. Despite the JAK/STAT pathway is recurrently altered in these neoplasms, there are not pharmacological inhibitors officially approved for the treatment of T-ALL/LBL patients that present oncogenic JAK/STAT pathway mutations. In the effort to identify potential therapeutic targets for those patients, we followed an alternative approach and focused on their transcriptional profile. We combined the analysis of molecular data from T-ALL/LBL patients with the generation of hematopoietic cellular models to reveal that JAK/STAT pathway mutations are associated with an aberrant transcriptional profile. Specifically, we demonstrate that JAK/STAT pathway mutations induce the overexpression of the PIM1 gene. Moreover, we show that the pan-PIM inhibitor, PIM447, significantly reduces the leukemogenesis, as well as the aberrant activation of c-MYC and mTOR pathways in cells expressing different JAK/STAT pathway mutations, becoming a potential therapeutic opportunity for a relevant subset of T-ALL/LBL patients.
Collapse
Affiliation(s)
- Antonio Lahera
- Department of Biology, Universidad Autónoma de Madrid, Madrid, 28049, Spain.
- Department of Genome dynamics and function, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28049, Spain.
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, Madrid, 28040, Spain.
| | - Laura Vela-Martín
- Department of Biology, Universidad Autónoma de Madrid, Madrid, 28049, Spain
- Department of Genome dynamics and function, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28049, Spain
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, Madrid, 28040, Spain
| | - Pablo Fernández-Navarro
- Unit of Cancer and Environmental Epidemiology, Centro Nacional de Epidemiología, Instituto de Salud Carlos III, Madrid, 28029, Spain
- Consorcio de Investigación Biomédica de Epidemiología y Salud Pública (CIBERESP), Madrid, 28029, Spain
| | - Pilar Llamas
- Division of Hematology and Hemotherapy, Hospital Universitario Fundación Jiménez Díaz, Madrid, 28040, Spain
| | - José L López-Lorenzo
- Division of Hematology and Hemotherapy, Hospital Universitario Fundación Jiménez Díaz, Madrid, 28040, Spain
| | - Javier Cornago
- Division of Hematology and Hemotherapy, Hospital Universitario Fundación Jiménez Díaz, Madrid, 28040, Spain
| | - Javier Santos
- Department of Biology, Universidad Autónoma de Madrid, Madrid, 28049, Spain
- Department of Genome dynamics and function, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28049, Spain
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, Madrid, 28040, Spain
- Institute for Molecular Biology-IUBM (Universidad Autónoma de Madrid) Madrid, Madrid, 28049, Spain
| | - José Fernández-Piqueras
- Department of Biology, Universidad Autónoma de Madrid, Madrid, 28049, Spain.
- Department of Genome dynamics and function, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28049, Spain.
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, Madrid, 28040, Spain.
- Institute for Molecular Biology-IUBM (Universidad Autónoma de Madrid) Madrid, Madrid, 28049, Spain.
| | - María Villa-Morales
- Department of Biology, Universidad Autónoma de Madrid, Madrid, 28049, Spain.
- Department of Genome dynamics and function, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28049, Spain.
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, Madrid, 28040, Spain.
- Institute for Molecular Biology-IUBM (Universidad Autónoma de Madrid) Madrid, Madrid, 28049, Spain.
| |
Collapse
|
2
|
Sumaria N, Fiala GJ, Inácio D, Curado-Avelar M, Cachucho A, Pinheiro R, Wiesheu R, Kimura S, Courtois L, Blankenhaus B, Darrigues J, Suske T, Almeida ARM, Minguet S, Asnafi V, Lhermitte L, Mullighan CG, Coffelt SB, Moriggl R, Barata JT, Pennington DJ, Silva-Santos B. Perinatal thymic-derived CD8αβ-expressing γδ T cells are innate IFN-γ producers that expand in IL-7R-STAT5B-driven neoplasms. Nat Immunol 2024; 25:1207-1217. [PMID: 38802512 PMCID: PMC11224017 DOI: 10.1038/s41590-024-01855-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/25/2024] [Indexed: 05/29/2024]
Abstract
The contribution of γδ T cells to immune responses is associated with rapid secretion of interferon-γ (IFN-γ). Here, we show a perinatal thymic wave of innate IFN-γ-producing γδ T cells that express CD8αβ heterodimers and expand in preclinical models of infection and cancer. Optimal CD8αβ+ γδ T cell development is directed by low T cell receptor signaling and through provision of interleukin (IL)-4 and IL-7. This population is pathologically relevant as overactive, or constitutive, IL-7R-STAT5B signaling promotes a supraphysiological accumulation of CD8αβ+ γδ T cells in the thymus and peripheral lymphoid organs in two mouse models of T cell neoplasia. Likewise, CD8αβ+ γδ T cells define a distinct subset of human T cell acute lymphoblastic leukemia pediatric patients. This work characterizes the normal and malignant development of CD8αβ+ γδ T cells that are enriched in early life and contribute to innate IFN-γ responses to infection and cancer.
Collapse
MESH Headings
- Animals
- Interferon-gamma/metabolism
- Interferon-gamma/immunology
- Mice
- Humans
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Thymus Gland/immunology
- Receptors, Interleukin-7/metabolism
- Immunity, Innate
- STAT5 Transcription Factor/metabolism
- Signal Transduction/immunology
- Mice, Inbred C57BL
- CD8-Positive T-Lymphocytes/immunology
- Mice, Knockout
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- CD8 Antigens/metabolism
- Female
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/metabolism
- Interleukin-7/metabolism
Collapse
Affiliation(s)
- Nital Sumaria
- Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Gina J Fiala
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
- Faculty of Biology, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| | - Daniel Inácio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Marta Curado-Avelar
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Cachucho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rúben Pinheiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Robert Wiesheu
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Lucien Courtois
- Hôpital Necker Enfants-Malades, Université de Paris, Paris, France
| | - Birte Blankenhaus
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Julie Darrigues
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tobias Suske
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Afonso R M Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Vahid Asnafi
- Hôpital Necker Enfants-Malades, Université de Paris, Paris, France
| | | | | | - Seth B Coffelt
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Richard Moriggl
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Daniel J Pennington
- Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK.
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
3
|
Dufva O. Unlocking a genetic model of NK-cell transformation. Blood 2024; 143:2443-2444. [PMID: 38869919 DOI: 10.1182/blood.2024024550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
|
4
|
Klein K, Kollmann S, Hiesinger A, List J, Kendler J, Klampfl T, Rhandawa M, Trifinopoulos J, Maurer B, Grausenburger R, Betram CA, Moriggl R, Rülicke T, Mullighan CG, Witalisz-Siepracka A, Walter W, Hoermann G, Sexl V, Gotthardt D. A lineage-specific STAT5BN642H mouse model to study NK-cell leukemia. Blood 2024; 143:2474-2489. [PMID: 38498036 PMCID: PMC11208297 DOI: 10.1182/blood.2023022655] [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: 09/22/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024] Open
Abstract
ABSTRACT Patients with T- and natural killer (NK)-cell neoplasms frequently have somatic STAT5B gain-of-function mutations. The most frequent STAT5B mutation is STAT5BN642H, which is known to drive murine T-cell leukemia, although its role in NK-cell malignancies is unclear. Introduction of the STAT5BN642H mutation into human NK-cell lines enhances their potential to induce leukemia in mice. We have generated a mouse model that enables tissue-specific expression of STAT5BN642H and have selectively expressed the mutated STAT5B in hematopoietic cells (N642Hvav/+) or exclusively in NK cells (N642HNK/NK). All N642Hvav/+ mice rapidly develop an aggressive T/NKT-cell leukemia, whereas N642HNK/NK mice display an indolent NK-large granular lymphocytic leukemia (NK-LGLL) that progresses to an aggressive leukemia with age. Samples from patients with NK-cell leukemia have a distinctive transcriptional signature driven by mutant STAT5B, which overlaps with that of murine leukemic N642HNK/NK NK cells. To our knowledge, we have generated the first reliable STAT5BN642H-driven preclinical mouse model that displays an indolent NK-LGLL progressing to aggressive NK-cell leukemia. This novel in vivo tool will enable us to explore the transition from an indolent to an aggressive disease and will thus permit the study of prevention and treatment options for NK-cell malignancies.
Collapse
Affiliation(s)
- Klara Klein
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Sebastian Kollmann
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Angela Hiesinger
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Julia List
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Jonatan Kendler
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thorsten Klampfl
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mehak Rhandawa
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Jana Trifinopoulos
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Barbara Maurer
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Reinhard Grausenburger
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Christof A. Betram
- Department for Biological Sciences and Pathobiology, Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Richard Moriggl
- Department for Biological Sciences and Pathobiology, Animal Breeding and Genetics, Unit for Functional Cancer Genomics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Rülicke
- Department for Biological Sciences and Pathobiology and Ludwig Boltzmann Institute for Hematology and Oncology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Charles G. Mullighan
- Department of Pathology, Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN
| | - Agnieszka Witalisz-Siepracka
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
- Division Pharmacology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | | | | | - Veronika Sexl
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
- University of Innsbruck, Innsbruck, Austria
| | - Dagmar Gotthardt
- Department for Biological Sciences and Pathobiology, Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
5
|
Dong Q, Wang Y, Xiu Y, Wu X, O’Neill S, Meyerson H, Suske T, Moriggl R, Hu S, Wang W, Zhao C. Unveiling myeloid transformation: T-LGLL with eosinophilia masking myeloid-associated STAT5B mutation culminating in AML. Br J Haematol 2024; 204:2487-2491. [PMID: 38508872 PMCID: PMC11178439 DOI: 10.1111/bjh.19421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Affiliation(s)
- Qianze Dong
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Yang Wang
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Yan Xiu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Xiaogang Wu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Stacey O’Neill
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio 44106, USA
| | - Howard Meyerson
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio 44106, USA
| | - Tobias Suske
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria
| | - Shimin Hu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Chen Zhao
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio 44106, USA
- Department of Medicine, Section of Hematology, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, USA
| |
Collapse
|
6
|
Yin CC, Tam W, Walker SM, Kaur A, Ouseph MM, Xie W, K Weinberg O, Li P, Zuo Z, Routbort MJ, Chen S, Medeiros LJ, George TI, Orazi A, Arber DA, Bagg A, Hasserjian RP, Wang SA. STAT5B mutations in myeloid neoplasms differ by disease subtypes but characterize a subset of chronic myeloid neoplasms with eosinophilia and/or basophilia. Haematologica 2024; 109:1825-1835. [PMID: 37981812 PMCID: PMC11141669 DOI: 10.3324/haematol.2023.284311] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/09/2023] [Indexed: 11/21/2023] Open
Abstract
STAT5B has been reported as a recurrent mutation in myeloid neoplasms with eosinophilia, but its overall frequency and importance across a spectrum of myeloid neoplasms are largely unknown. We conducted a multicenter study on a series of 82 myeloid neoplasms with STAT5B mutations detected by next-generation sequencing. The estimated frequency of STAT5B mutations in myeloid neoplasms was low, <0.5%, but mutations were detected in all categories of such neoplasms, including myelodysplastic syndrome (MDS, 28%), acute myeloid leukemia (AML, 26%), myelodysplastic/myeloproliferative neoplasm (MDS/MPN, 18%), Philadelphia chromosome-negative classic MPN (12%), systemic mastocytosis (1%), and, with a notably high frequency, chronic eosinophilic leukemia, not otherwise specified (CEL-NOS, 15%). STAT5B mutations occurred preferentially in the SH2 domain (95%), involved 12 different codons, with the N642H hotspot being the most common (78%). Co-mutations were present in all cases and clonal hierarchy analysis showed that STAT5B mutations tended to be subclonal in AML, MPN, and MDS, but frequently dominant/co-dominant in CEL-NOS (83%), followed by MDS/MPN (40%). Across the group, eosinophilia and/or basophilia were common (41%), frequently observed in cases in which STAT5B mutations were detected at initial diagnosis (P<0.0001), with a high variant allele frequency (median 42.5%, P=0.0001), as a dominant/ co-dominant clone (P<0.0001), involving the canonical N642H (P=0.0607), and associated with fewer co-mutations (P=0.0009). Our data show that the characteristics and importance of a STAT5B mutation differ among myeloid neoplasms, but if present as a dominant mutation and detected at initial diagnosis, it appears to be a driver mutation in a subgroup of chronic myeloid neoplasms, preferentially promoting a proliferation of eosinophils and basophils.
Collapse
Affiliation(s)
- C Cameron Yin
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Wayne Tam
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Greenvale, NY
| | - Serena M Walker
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Amandeep Kaur
- Department of Pathology, University of Chicago, Chicago, IL
| | - Madhu M Ouseph
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical Center, New York, NY
| | - Wei Xie
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR
| | - Olga K Weinberg
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Peng Li
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Zhuang Zuo
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mark J Routbort
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Simon Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - L Jeffrey Medeiros
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tracy I George
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Attilio Orazi
- Department of Pathology, Texas Tech University, El Paso, TX
| | - Daniel A Arber
- Department of Pathology, University of Chicago, Chicago, IL
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Sa A Wang
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
7
|
Fortelny N, Farlik M, Fife V, Gorki AD, Lassnig C, Maurer B, Meissl K, Dolezal M, Boccuni L, Ravi Sundar Jose Geetha A, Akagha MJ, Karjalainen A, Shoebridge S, Farhat A, Mann U, Jain R, Tikoo S, Zila N, Esser-Skala W, Krausgruber T, Sitnik K, Penz T, Hladik A, Suske T, Zahalka S, Senekowitsch M, Barreca D, Halbritter F, Macho-Maschler S, Weninger W, Neubauer HA, Moriggl R, Knapp S, Sexl V, Strobl B, Decker T, Müller M, Bock C. JAK-STAT signaling maintains homeostasis in T cells and macrophages. Nat Immunol 2024; 25:847-859. [PMID: 38658806 PMCID: PMC11065702 DOI: 10.1038/s41590-024-01804-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 03/07/2024] [Indexed: 04/26/2024]
Abstract
Immune cells need to sustain a state of constant alertness over a lifetime. Yet, little is known about the regulatory processes that control the fluent and fragile balance that is called homeostasis. Here we demonstrate that JAK-STAT signaling, beyond its role in immune responses, is a major regulator of immune cell homeostasis. We investigated JAK-STAT-mediated transcription and chromatin accessibility across 12 mouse models, including knockouts of all STAT transcription factors and of the TYK2 kinase. Baseline JAK-STAT signaling was detected in CD8+ T cells and macrophages of unperturbed mice-but abrogated in the knockouts and in unstimulated immune cells deprived of their normal tissue context. We observed diverse gene-regulatory programs, including effects of STAT2 and IRF9 that were independent of STAT1. In summary, our large-scale dataset and integrative analysis of JAK-STAT mutant and wild-type mice uncovered a crucial role of JAK-STAT signaling in unstimulated immune cells, where it contributes to a poised epigenetic and transcriptional state and helps prepare these cells for rapid response to immune stimuli.
Collapse
Affiliation(s)
- Nikolaus Fortelny
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Tumor Biology and Immunology, Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| | - Victoria Fife
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Anna-Dorothea Gorki
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Caroline Lassnig
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Barbara Maurer
- Pharmacology and Toxicology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Katrin Meissl
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Marlies Dolezal
- Platform for Bioinformatics and Biostatistics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Laura Boccuni
- Max Perutz Labs, University of Vienna, Vienna, Austria
| | | | - Mojoyinola Joanna Akagha
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Anzhelika Karjalainen
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Stephen Shoebridge
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Asma Farhat
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Ulrike Mann
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Rohit Jain
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Shweta Tikoo
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Nina Zila
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Esser-Skala
- Center for Tumor Biology and Immunology, Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Katarzyna Sitnik
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Thomas Penz
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Anastasiya Hladik
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Tobias Suske
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Sophie Zahalka
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Martin Senekowitsch
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Daniele Barreca
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Florian Halbritter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Sabine Macho-Maschler
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Heidi A Neubauer
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Richard Moriggl
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Sylvia Knapp
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Veronika Sexl
- Pharmacology and Toxicology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
- University of Innsbruck, Innsbruck, Austria
| | - Birgit Strobl
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Thomas Decker
- Max Perutz Labs, University of Vienna, Vienna, Austria
| | - Mathias Müller
- Animal Breeding and Genetics and VetBiomodels, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
8
|
Suske T, Sorger H, Manhart G, Ruge F, Prutsch N, Zimmerman MW, Eder T, Abdallah DI, Maurer B, Wagner C, Schönefeldt S, Spirk K, Pichler A, Pemovska T, Schweicker C, Pölöske D, Hubanic E, Jungherz D, Müller TA, Aung MMK, Orlova A, Pham HTT, Zimmel K, Krausgruber T, Bock C, Müller M, Dahlhoff M, Boersma A, Rülicke T, Fleck R, de Araujo ED, Gunning PT, Aittokallio T, Mustjoki S, Sanda T, Hartmann S, Grebien F, Hoermann G, Haferlach T, Staber PB, Neubauer HA, Look AT, Herling M, Moriggl R. Hyperactive STAT5 hijacks T cell receptor signaling and drives immature T cell acute lymphoblastic leukemia. J Clin Invest 2024; 134:e168536. [PMID: 38618957 PMCID: PMC11014662 DOI: 10.1172/jci168536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/27/2024] [Indexed: 04/16/2024] Open
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive immature T cell cancer. Mutations in IL7R have been analyzed genetically, but downstream effector functions such as STAT5A and STAT5B hyperactivation are poorly understood. Here, we studied the most frequent and clinically challenging STAT5BN642H driver in T cell development and immature T cell cancer onset and compared it with STAT5A hyperactive variants in transgenic mice. Enhanced STAT5 activity caused disrupted T cell development and promoted an early T cell progenitor-ALL phenotype, with upregulation of genes involved in T cell receptor (TCR) signaling, even in absence of surface TCR. Importantly, TCR pathway genes were overexpressed in human T-ALL and mature T cell cancers and activation of TCR pathway kinases was STAT5 dependent. We confirmed STAT5 binding to these genes using ChIP-Seq analysis in human T-ALL cells, which were sensitive to pharmacologic inhibition by dual STAT3/5 degraders or ZAP70 tyrosine kinase blockers in vitro and in vivo. We provide genetic and biochemical proof that STAT5A and STAT5B hyperactivation can initiate T-ALL through TCR pathway hijacking and suggest similar mechanisms for other T cell cancers. Thus, STAT5 or TCR component blockade are targeted therapy options, particularly in patients with chemoresistant clones carrying STAT5BN642H.
Collapse
Affiliation(s)
| | | | - Gabriele Manhart
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Frank Ruge
- Institute of Animal Breeding and Genetics and
| | - Nicole Prutsch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark W. Zimmerman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Eder
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Diaaeldin I. Abdallah
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | - Alexander Pichler
- Department of Medicine I, Clinical Division of Hematology, Medical University of Vienna, Vienna, Austria
| | - Tea Pemovska
- Department of Medicine I, Clinical Division of Hematology, Medical University of Vienna, Vienna, Austria
| | - Carmen Schweicker
- Department of Medicine I, Clinical Division of Hematology, Medical University of Vienna, Vienna, Austria
| | | | | | - Dennis Jungherz
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, University of Cologne, Cologne, Germany
| | - Tony Andreas Müller
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, University of Cologne, Cologne, Germany
| | | | - Anna Orlova
- Institute of Animal Breeding and Genetics and
| | | | | | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | | | - Maik Dahlhoff
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Auke Boersma
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Rülicke
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Elvin Dominic de Araujo
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Patrick Thomas Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Janpix, London, United Kingdom
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Satu Mustjoki
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Takaomi Sanda
- Cancer Science Institute of Singapore and Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of Pathology, Goethe University, Frankfurt am Main, Germany
| | - Florian Grebien
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | | | | | - Philipp Bernhard Staber
- Department of Medicine I, Clinical Division of Hematology, Medical University of Vienna, Vienna, Austria
| | | | - Alfred Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Marco Herling
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, University of Cologne, Cologne, Germany
- Department of Hematology, Cellular Therapy and Hemostaseology, University of Leipzig, Leipzig, Germany
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics and
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| |
Collapse
|
9
|
Liongue C, Ratnayake T, Basheer F, Ward AC. Janus Kinase 3 (JAK3): A Critical Conserved Node in Immunity Disrupted in Immune Cell Cancer and Immunodeficiency. Int J Mol Sci 2024; 25:2977. [PMID: 38474223 DOI: 10.3390/ijms25052977] [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: 01/17/2024] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
The Janus kinase (JAK) family is a small group of protein tyrosine kinases that represent a central component of intracellular signaling downstream from a myriad of cytokine receptors. The JAK3 family member performs a particularly important role in facilitating signal transduction for a key set of cytokine receptors that are essential for immune cell development and function. Mutations that impact JAK3 activity have been identified in a number of human diseases, including somatic gain-of-function (GOF) mutations associated with immune cell malignancies and germline loss-of-function (LOF) mutations associated with immunodeficiency. The structure, function and impacts of both GOF and LOF mutations of JAK3 are highly conserved, making animal models highly informative. This review details the biology of JAK3 and the impact of its perturbation in immune cell-related diseases, including relevant animal studies.
Collapse
Affiliation(s)
- Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3216, Australia
| | | | - Faiza Basheer
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3216, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3216, Australia
| |
Collapse
|
10
|
Liang D, Wang Q, Zhang W, Tang H, Song C, Yan Z, Liang Y, Wang H. JAK/STAT in leukemia: a clinical update. Mol Cancer 2024; 23:25. [PMID: 38273387 PMCID: PMC10811937 DOI: 10.1186/s12943-023-01929-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
Over the past three decades, considerable efforts have been expended on understanding the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway in leukemia, following the identification of the JAK2V617F mutation in myeloproliferative neoplasms (MPNs). The aim of this review is to summarize the latest progress in our understanding of the involvement of the JAK/STAT signaling pathway in the development of leukemia. We also attempt to provide insights into the current use of JAK/STAT inhibitors in leukemia therapy and explore pertinent clinical trials in this field.
Collapse
Affiliation(s)
- Dong Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qiaoli Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wenbiao Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Cailu Song
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhimin Yan
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
| | - Yang Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Hua Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
11
|
Swain RM, Sanchez A, Gutierrez DA, Varela-Ramirez A, Aguilera RJ. Thiophene derivative inflicts cytotoxicity via an intrinsic apoptotic pathway on human acute lymphoblastic leukemia cells. PLoS One 2023; 18:e0295441. [PMID: 38127921 PMCID: PMC10734950 DOI: 10.1371/journal.pone.0295441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
In an effort to identify novel anti-cancer agents, we employed a well-established High Throughput Screening (HTS) assay to assess the cytotoxic effect of compounds within the ChemBridge DIVERSet Library on a lymphoma cell line. This screen revealed a novel thiophene, F8 (methyl 5-[(dimethylamino)carbonyl]-4-methyl-2-[(3-phenyl-2-propynoyl) amino]-3-thiophenecarboxylate), that displays anti-cancer activity on lymphoma, leukemia, and other cancer cell lines. Thiophenes and thiophene derivatives have emerged as an important class of heterocyclic compounds that have displayed favorable drug characteristics. They have been previously reported to exhibit a broad spectrum of properties and varied uses in the field of medicine. In addition, they have proven to be effective drugs in various disease scenarios. They contain anti-inflammatory, anti-anxiety, anti-psychotic, anti-microbial, anti-fungal, estrogen receptor modulating, anti-mitotic, kinase inhibiting and anti-cancer activities, rendering compounds with a thiophene a subject of significant interest in the scientific community. Compound F8 consistently induced cell death at a low micromolar range on a small panel of cancer cell lines after a 48 h period. Further investigation revealed that F8 induced phosphatidylserine externalization, reactive oxygen species generation, mitochondrial depolarization, kinase inhibition, and induces apoptosis. These findings demonstrate that F8 has promising anti-cancer activity.
Collapse
Affiliation(s)
- Risa Mia Swain
- Department of Biological Sciences, The Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, United States of America
- Department of Molecular and Translational Medicine, Center of Emphasis in Cancer, Paul Foster School of Medicine, Texas Tech University Health Science Center El Paso, El Paso, Texas, United States of America
| | - Anahi Sanchez
- Department of Biological Sciences, The Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, United States of America
| | - Denisse A. Gutierrez
- Department of Biological Sciences, The Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, United States of America
| | - Armando Varela-Ramirez
- Department of Biological Sciences, The Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, United States of America
| | - Renato J. Aguilera
- Department of Biological Sciences, The Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, United States of America
| |
Collapse
|
12
|
Li J, Tang B, Miao Y, Li G, Sun Z. Targeting of STAT5 using the small molecule topotecan hydrochloride suppresses acute myeloid leukemia progression. Oncol Rep 2023; 50:208. [PMID: 37830151 PMCID: PMC10603551 DOI: 10.3892/or.2023.8645] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
Acute myeloid leukemia (AML) is a common type of acute leukemia in adults and relapse is one of the main reasons for treatment failure. FLT3‑ITD mutations are associated with poor prognosis, short disease‑free progression survival and high relapse rates in patients with AML. STAT5 is activated by FLT3‑ITD and drives the pathogenesis of AML. STAT5 activation is usually a hallmark of hematologic malignancies and occurs in ~70% of patients with AML. Moreover, STAT5 is a key molecule which regulates hematopoiesis, and its high expression is closely associated with drug resistance, thus direct targeting of STAT5 for AML is of great clinical value. The present study introduces a new small‑molecule inhibitor that targets STAT5, presenting a promising approach for AML therapy. A high throughput fluorescence polarization (FP) screening system for STAT5 was designed and established, and used to screen an existing compound library to obtain the highly active small molecule inhibitor, topotecan hydrochloride. Topotecan hydrochloride was demonstrated to be an effective inhibitor of STAT5 by molecular docking prediction and cellular thermal shift assay. Topotecan hydrochloride bound to STAT5, inhibiting its dimerization, phosphorylation and transcription of specific target genes. The compound exhibits cellular activity at the nanomolar level and significantly inhibits the proliferation of human AML cell lines and FLT3‑ITD+ AML cells. Furthermore, topotecan hydrochloride has the potential to exert an anti‑tumor effect in vivo. Overall, topotecan hydrochloride offers a new opportunity for the treatment of AML and other hematologic malignancies by directly targeting STAT5.
Collapse
Affiliation(s)
- Jiahui Li
- Fengxian Hospital Affiliated to Anhui University of Science and Technology, Shanghai 201499, P.R. China
| | - Bin Tang
- Department of Gynecology, East China Normal University Wuhu Affiliated Hospital (The Second People's Hospital of Wuhu City), Wuhu, Anhui 241000, P.R. China
| | - Ying Miao
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Center for Translational Medicine, Shanghai Key Laboratory of Regulatory Biology Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 201100, P.R. China
| | - Guihong Li
- Fengxian Hospital Affiliated to The Southern Medical University, Shanghai 201499, P.R. China
| | - Zhenliang Sun
- Fengxian Hospital Affiliated to Anhui University of Science and Technology, Shanghai 201499, P.R. China
| |
Collapse
|
13
|
Kobets AJ, Ahmad S, Boyke A, Oriko D, Holland R, Eisenberg R, Alavi SAN, Abbott R. STAT5b gain-of-function disease in a child with mycobacterial osteomyelitis of the skull: rare presentation of an emerging disease entity. Childs Nerv Syst 2023:10.1007/s00381-023-05997-y. [PMID: 37243811 DOI: 10.1007/s00381-023-05997-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/18/2023] [Indexed: 05/29/2023]
Abstract
PURPOSE STAT proteins play a key role in several cellular functions related to cell development, differentiation, proliferation, and survival. Persistent STAT activation due to somatic STAT5bN642H gain-of-function mutation is a rare mechanism of STAT dysregulation that results in hypereosinophilia, frequent infections, leukemias, and pulmonary diseases. Herein, we describe a case of a child with a rare early onset STAT5b gain-of-function disease treated with targeted JAK inhibition who developed a cranial Mycobacterium avium osteomyelitis. METHODS A 3-year-old male with a known STAT5b gain-of-function mutation presented with a 10-day history of a firm, immobile, non-painful cranial mycobacterium mass with dural infiltration located anterior to the coronal suture. Stepwise management finalized with complete resection of the lesion with calvarial reconstruction. A case-based literature review was performed evaluating all patients with this mutation who developed cranial disease. RESULTS The patient was symptom and lesion-free at 1 year since surgical resection and initiation of triple mycobacterial pharmacotherapy. Our literature review demonstrated the rarity of this disease, as well as other presentations of this disease in other patients. CONCLUSION Patients with STAT5b gain-of-function mutations have attenuated Th1 responses and are treated with medications, such as JAK inhibitors, which further inhibit other STAT proteins that regulate immunity against rare infectious entities, such as mycobacterium. Our case highlights the importance of considering these rare infections in patients on JAK inhibitors and with STAT protein mutations. Possessing a clear mechanistic understanding of this genetic mutation, its downstream effect, and the consequences of treatment may enhance a physician's diagnostic and clinical management of similar patients in the future.
Collapse
Affiliation(s)
- Andrew J Kobets
- Department of Neurosurgery, Montefiore Medical Center and the Albert Einstein College of Medicine, 3316 Rochambeau Ave, Bronx, NY, 10467, USA.
| | - Samuel Ahmad
- Department of Neurosurgery, Montefiore Medical Center and the Albert Einstein College of Medicine, 3316 Rochambeau Ave, Bronx, NY, 10467, USA
| | - Andre Boyke
- Department of Neurosurgery, Montefiore Medical Center and the Albert Einstein College of Medicine, 3316 Rochambeau Ave, Bronx, NY, 10467, USA
| | - David Oriko
- University of Nairobi School of Medicine, Nairobi, Kenya
| | - Ryan Holland
- Department of Neurosurgery, Montefiore Medical Center and the Albert Einstein College of Medicine, 3316 Rochambeau Ave, Bronx, NY, 10467, USA
| | - Rachel Eisenberg
- Department of Pediatrics, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Rick Abbott
- Department of Neurosurgery, Montefiore Medical Center and the Albert Einstein College of Medicine, 3316 Rochambeau Ave, Bronx, NY, 10467, USA
| |
Collapse
|
14
|
Van der Zwet JCG, Cordo' V, Buijs-Gladdines JGCAM, Hagelaar R, Smits WK, Vroegindeweij E, Graus LTM, Poort V, Nulle M, Pieters R, Meijerink JPP. STAT5 does not drive steroid resistance in T-cell acute lymphoblastic leukemia despite the activation of BCL2 and BCLXL following glucocorticoid treatment. Haematologica 2023; 108:732-746. [PMID: 35734930 PMCID: PMC9973477 DOI: 10.3324/haematol.2021.280405] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Indexed: 11/09/2022] Open
Abstract
Physiological and pathogenic interleukin-7-receptor (IL7R)-induced signaling provokes glucocorticoid resistance in a subset of patients with pediatric T-cell acute lymphoblastic leukemia (T-ALL). Activation of downstream STAT5 has been suggested to cause steroid resistance through upregulation of anti-apoptotic BCL2, one of its downstream target genes. Here we demonstrate that isolated STAT5 signaling in various T-ALL cell models is insufficient to raise cellular steroid resistance despite upregulation of BCL2 and BCL-XL. Upregulation of anti-apoptotic BCL2 and BCLXL in STAT5-activated T-ALL cells requires steroid-induced activation of NR3C1. For the BCLXL locus, this is facilitated by a concerted action of NR3C1 and activated STAT5 molecules at two STAT5 regulatory sites, whereas for the BCL2 locus this is facilitated by binding of NR3C1 at a STAT5 binding motif. In contrast, STAT5 occupancy at glucocorticoid response elements does not affect the expression of NR3C1 target genes. Strong upregulation of BIM, a NR3C1 pro-apoptotic target gene, upon prednisolone treatment can counterbalance NR3C1/STAT5-induced BCL2 and BCL-XL expression downstream of IL7- induced or pathogenic IL7R signaling. This explains why isolated STAT5 activation does not directly impair the steroid response. Our study suggests that STAT5 activation only contributes to steroid resistance in combination with cellular defects or alternative signaling routes that disable the pro-apoptotic and steroid-induced BIM response.
Collapse
Affiliation(s)
| | | | | | - Rico Hagelaar
- Princess Maxima Center for Pediatric Oncology, Utrecht
| | | | | | | | - Vera Poort
- Princess Maxima Center for Pediatric Oncology, Utrecht
| | - Marloes Nulle
- Princess Maxima Center for Pediatric Oncology, Utrecht
| | - Rob Pieters
- Princess Maxima Center for Pediatric Oncology, Utrecht
| | | |
Collapse
|
15
|
Pentacyclic Triterpenoids-Based Ionic Compounds: Synthesis, Study of Structure-Antitumor Activity Relationship, Effects on Mitochondria and Activation of Signaling Pathways of Proliferation, Genome Reparation and Early Apoptosis. Cancers (Basel) 2023; 15:cancers15030756. [PMID: 36765714 PMCID: PMC9913425 DOI: 10.3390/cancers15030756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/16/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
The present research paper details the synthesis of novel ionic compounds based on triterpene acids (betulinic, oleanolic and ursolic), with these acids acting both as anions and connected through a spacer with various nitrogen-containing compounds (pyridine, piperidine, morpholine, pyrrolidine, triethylamine and dimethylethanolamine) and acting as a cation. Based on the latter, a large number of ionic compounds with various counterions (BF4-, SbF6-, PF6-, CH3COO-, C6H5SO3-, m-C6H4(OH)COO- and CH3CH(OH)COO-) have been synthesized. We studied the cytotoxicity of the synthesized compounds on the example of various tumor (Jurkat, K562, U937, HL60, A2780) and conditionally normal (HEK293) cell lines. IC50 was determined, and the influence of the structure and nature of the anion and cation on the antitumor activity was specified. Intracellular signaling, apoptosis induction and effects of the most active ionic compounds on the cell cycle and mitochondria have been discussed by applying modern methods of multiparametric enzyme immunoassay and flow cytometry.
Collapse
|
16
|
Umrau K, Naganuma K, Gao Q, Dogan A, Kizaki M, Roshal M, Liu Y, Yabe M. Activating STAT5B mutations can cause both primary hypereosinophilia and lymphocyte-variant hypereosinophilia. Leuk Lymphoma 2023; 64:238-241. [PMID: 36308018 PMCID: PMC11026062 DOI: 10.1080/10428194.2022.2131413] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/09/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Kavita Umrau
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ken Naganuma
- Department of Hematology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Qi Gao
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ahmet Dogan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Masahiro Kizaki
- Department of Hematology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Mikhail Roshal
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying Liu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mariko Yabe
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
17
|
Matza Porges S, Shamriz O. Genetics of Immune Dysregulation and Cancer Predisposition: Two Sides of the Same Coin. Clin Exp Immunol 2022; 210:114-127. [PMID: 36165533 PMCID: PMC9750831 DOI: 10.1093/cei/uxac089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/17/2022] [Accepted: 09/23/2022] [Indexed: 01/25/2023] Open
Abstract
Approximately 10% of cancers have a hereditary predisposition. However, no genetic diagnosis is available in 60%-80% of familial cancers. In some of these families, immune dysregulation-mediated disease is frequent. The immune system plays a critical role in identifying and eliminating tumors; thus, dysregulation of the immune system can increase the risk of developing cancer. This review focuses on some of the genes involved in immune dysregulation the promote the risk for cancer. Genetic counseling for patients with cancer currently focuses on known genes that raise the risk of cancer. In missing hereditary familial cases, the history family of immune dysregulation should be recorded, and genes related to the immune system should be analyzed in relevant families. On the other hand, patients with immune disorders diagnosed with a pathogenic mutation in an immune regulatory gene may have an increased risk of cancer. Therefore, those patients need to be under surveillance for cancer. Gene panel and exome sequencing are currently standard methods for genetic diagnosis, providing an excellent opportunity to jointly test cancer and immune genes.
Collapse
Affiliation(s)
- Sigal Matza Porges
- Department of Human Genetics, Institute for Medical Research, the Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Biotechnology, Hadassah Academic College, Jerusalem, Israel
| | - Oded Shamriz
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah Medical Organization, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
18
|
Zhang Z, Yang K, Zhang H. Targeting Leukemia-Initiating Cells and Leukemic Niches: The Next Therapy Station for T-Cell Acute Lymphoblastic Leukemia? Cancers (Basel) 2022; 14:cancers14225655. [PMID: 36428753 PMCID: PMC9688677 DOI: 10.3390/cancers14225655] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive subtype of hematological malignancy characterized by its high heterogeneity and potentially life-threatening clinical features. Despite the advances in risk stratification and therapeutic management of T-ALL, patients often suffer from treatment failure and chemotherapy-induced toxicity, calling for greater efforts to improve therapeutic efficacy and safety in the treatment of T-ALL. During the past decades, increasing evidence has shown the indispensable effects of leukemia-initiating cells (LICs) and leukemic niches on T-ALL initiation and progression. These milestones greatly facilitate precision medicine by interfering with the pathways that are associated with LICs and leukemic niches or by targeting themselves directly. Most of these novel agents, either alone or in combination with conventional chemotherapy, have shown promising preclinical results, facilitating them to be further evaluated under clinical trials. In this review, we summarize the latest discoveries in LICs and leukemic niches in terms of T-ALL, with a particular highlight on the current precision medicine. The challenges and future prospects are also discussed.
Collapse
Affiliation(s)
- Ziting Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Kun Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Correspondence: ; Tel.: +86-158-7796-3252
| |
Collapse
|
19
|
Garces de Los Fayos Alonso I, Zujo L, Wiest I, Kodajova P, Timelthaler G, Edtmayer S, Zrimšek M, Kollmann S, Giordano C, Kothmayer M, Neubauer HA, Dey S, Schlederer M, Schmalzbauer BS, Limberger T, Probst C, Pusch O, Högler S, Tangermann S, Merkel O, Schiefer AI, Kornauth C, Prutsch N, Zimmerman M, Abraham B, Anagnostopoulos J, Quintanilla-Martinez L, Mathas S, Wolf P, Stoiber D, Staber PB, Egger G, Klapper W, Woessmann W, Look TA, Gunning P, Turner SD, Moriggl R, Lagger S, Kenner L. PDGFRβ promotes oncogenic progression via STAT3/STAT5 hyperactivation in anaplastic large cell lymphoma. Mol Cancer 2022; 21:172. [PMID: 36045346 PMCID: PMC9434917 DOI: 10.1186/s12943-022-01640-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/31/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Anaplastic large cell lymphoma (ALCL) is an aggressive non-Hodgkin T cell lymphoma commonly driven by NPM-ALK. AP-1 transcription factors, cJUN and JUNb, act as downstream effectors of NPM-ALK and transcriptionally regulate PDGFRβ. Blocking PDGFRβ kinase activity with imatinib effectively reduces tumor burden and prolongs survival, although the downstream molecular mechanisms remain elusive. METHODS AND RESULTS In a transgenic mouse model that mimics PDGFRβ-driven human ALCL in vivo, we identify PDGFRβ as a driver of aggressive tumor growth. Mechanistically, PDGFRβ induces the pro-survival factor Bcl-xL and the growth-enhancing cytokine IL-10 via STAT5 activation. CRISPR/Cas9 deletion of both STAT5 gene products, STAT5A and STAT5B, results in the significant impairment of cell viability compared to deletion of STAT5A, STAT5B or STAT3 alone. Moreover, combined blockade of STAT3/5 activity with a selective SH2 domain inhibitor, AC-4-130, effectively obstructs tumor development in vivo. CONCLUSIONS We therefore propose PDGFRβ as a novel biomarker and introduce PDGFRβ-STAT3/5 signaling as an important axis in aggressive ALCL. Furthermore, we suggest that inhibition of PDGFRβ or STAT3/5 improve existing therapies for both previously untreated and relapsed/refractory ALK+ ALCL patients.
Collapse
Affiliation(s)
- I Garces de Los Fayos Alonso
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - L Zujo
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
- Division of Nuclear Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - I Wiest
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
- Division of Nuclear Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - P Kodajova
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - G Timelthaler
- Center for Cancer Research, Medical University of Vienna, 1090, Vienna, Austria
| | - S Edtmayer
- Division Pharmacology, Department of Pharmacology, Physiology and Microbiology, Karl Landsteiner University of Health Sciences, 3500, Krems, Austria
| | - M Zrimšek
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
| | - S Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - C Giordano
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
| | - M Kothmayer
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
- Centre for Anatomy and Cell Biology, Medical University of Vienna, 1090, Vienna, Austria
| | - H A Neubauer
- Institute of Animal Breeding and Genetics, Unit of Functional Cancer Genomics, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - S Dey
- Department of Dermatology, Medical University of Graz, 8036, Graz, Austria
- Center for Medical Research (ZMF), Medical University of Graz, 8010, Graz, Austria
| | - M Schlederer
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
| | - B S Schmalzbauer
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - T Limberger
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Division of Nuclear Medicine, Medical University of Vienna, 1090, Vienna, Austria
- CBMed Core Lab, Medical University of Vienna, 1090, Vienna, Austria
| | - C Probst
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
- Division of Nuclear Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - O Pusch
- Centre for Anatomy and Cell Biology, Medical University of Vienna, 1090, Vienna, Austria
| | - S Högler
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - S Tangermann
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - O Merkel
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
| | - A I Schiefer
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
| | - C Kornauth
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center Vienna, Vienna General Hospital, Medical University of Vienna, 1090, Vienna, Austria
| | - N Prutsch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - M Zimmerman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - B Abraham
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - J Anagnostopoulos
- Institute of Pathology, University of Wuerzburg, 97080, Würzburg, Germany
- Institute of Pathology, Charité-Medical University of Berlin, 10117, Berlin, Germany
| | - L Quintanilla-Martinez
- Institute of Pathology and Neuropathology and Cluster of excellence iFIT, "Image-Guided and Functionally Instructed Tumor Therapy", University of Tübingen, 72076, Tübingen, Germany
| | - S Mathas
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Medical University of Berlin, 12200, Berlin, Germany
- German Cancer Consortium (DKTK) German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125, Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125, Berlin, Germany
| | - P Wolf
- Department of Dermatology, Medical University of Graz, 8036, Graz, Austria
| | - D Stoiber
- Division Pharmacology, Department of Pharmacology, Physiology and Microbiology, Karl Landsteiner University of Health Sciences, 3500, Krems, Austria
| | - P B Staber
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center Vienna, Vienna General Hospital, Medical University of Vienna, 1090, Vienna, Austria
| | - G Egger
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center Vienna, Vienna General Hospital, Medical University of Vienna, 1090, Vienna, Austria
- Boltzmann Institute Applied Diagnostics, 1090, Vienna, Austria
| | - W Klapper
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University of Kiel/University Hospital Schleswig-Holstein, 24105, Kiel, Germany
| | - W Woessmann
- Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - T A Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - P Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON, L5L 1C6, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
| | - S D Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, CB20QQ, UK
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - R Moriggl
- Institute of Animal Breeding and Genetics, Unit of Functional Cancer Genomics, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - S Lagger
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - L Kenner
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria.
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria.
- Division of Nuclear Medicine, Medical University of Vienna, 1090, Vienna, Austria.
- Center for Medical Research (ZMF), Medical University of Graz, 8010, Graz, Austria.
- CBMed Core Lab, Medical University of Vienna, 1090, Vienna, Austria.
- Christian Doppler Laboratory of Applied Metabolomics, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
20
|
Concept of Hybrid Drugs and Recent Advancements in Anticancer Hybrids. Pharmaceuticals (Basel) 2022; 15:ph15091071. [PMID: 36145292 PMCID: PMC9500727 DOI: 10.3390/ph15091071] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is a complex disease, and its treatment is a big challenge, with variable efficacy of conventional anticancer drugs. A two-drug cocktail hybrid approach is a potential strategy in recent drug discovery that involves the combination of two drug pharmacophores into a single molecule. The hybrid molecule acts through distinct modes of action on several targets at a given time with more efficacy and less susceptibility to resistance. Thus, there is a huge scope for using hybrid compounds to tackle the present difficulties in cancer medicine. Recent work has applied this technique to uncover some interesting molecules with substantial anticancer properties. In this study, we report data on numerous promising hybrid anti-proliferative/anti-tumor agents developed over the previous 10 years (2011–2021). It includes quinazoline, indole, carbazole, pyrimidine, quinoline, quinone, imidazole, selenium, platinum, hydroxamic acid, ferrocene, curcumin, triazole, benzimidazole, isatin, pyrrolo benzodiazepine (PBD), chalcone, coumarin, nitrogen mustard, pyrazole, and pyridine-based anticancer hybrids produced via molecular hybridization techniques. Overall, this review offers a clear indication of the potential benefits of merging pharmacophoric subunits from multiple different known chemical prototypes to produce more potent and precise hybrid compounds. This provides valuable knowledge for researchers working on complex diseases such as cancer.
Collapse
|
21
|
Zhang Z, Salgado OC, Liu B, Moazzami Z, Hogquist KA, Farrar MA, Ruan HB. An OGT-STAT5 Axis in Regulatory T Cells Controls Energy and Iron Metabolism. Front Immunol 2022; 13:874863. [PMID: 35874700 PMCID: PMC9304952 DOI: 10.3389/fimmu.2022.874863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
The immunosuppressive regulatory T (Treg) cells exert emerging effects on adipose tissue homeostasis and systemic metabolism. However, the metabolic regulation and effector mechanisms of Treg cells in coping with obesogenic insults are not fully understood. We have previously established an indispensable role of the O-linked N-Acetylglucosamine (O-GlcNAc) signaling in maintaining Treg cell identity and promoting Treg suppressor function, via STAT5 O-GlcNAcylation and activation. Here, we investigate the O-GlcNAc transferase (OGT)-STAT5 axis in driving the immunomodulatory function of Treg cells for metabolic homeostasis. Treg cell-specific OGT deficiency renders mice more vulnerable to high-fat diet (HFD)-induced adiposity and insulin resistance. Conversely, constitutive STAT5 activation in Treg cells confers protection against adipose tissue expansion and impaired glucose and insulin metabolism upon HFD feeding, in part by suppressing adipose lipid uptake and redistributing systemic iron storage. Treg cell function can be augmented by targeting the OGT-STAT5 axis to combat obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Zengdi Zhang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Oscar C. Salgado
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Bing Liu
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Zahra Moazzami
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
- Department of Food Science and Nutrition, University of Minnesota, Minneapolis, MN, United States
| | - Kristin A. Hogquist
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Michael A. Farrar
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Hai-Bin Ruan,
| |
Collapse
|
22
|
Veloza L, Cavalieri D, Missiaglia E, Ledoux-Pilon A, Bisig B, Pereira B, Bonnet C, Poullot E, Quintanilla-Martinez L, Dubois R, Llamas-Gutierrez F, Bossard C, De Wind R, Drieux F, Fontaine J, Parrens M, Sandrini J, Fataccioli V, Delfau-Larue MH, Daniel A, Lhomme F, Clément-Filliatre L, Lemonnier F, Cairoli A, Morel P, Glaisner S, Joly B, El Yamani A, Laribi K, Bachy E, Siebert R, Vallois D, Gaulard P, Tournilhac O, de Leval L. Monomorphic epitheliotropic intestinal T-cell lymphoma comprises morphologic and genomic heterogeneity impacting outcome. Haematologica 2022; 108:181-195. [PMID: 35708139 PMCID: PMC9827163 DOI: 10.3324/haematol.2022.281226] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Indexed: 02/05/2023] Open
Abstract
Monomorphic epitheliotropic intestinal T-cell lymphoma (MEITL) is a rare aggressive T-cell lymphoma most reported in Asia. We performed a comprehensive clinical, pathological and genomic study of 71 European MEITL patients (36 males, 35 females, median age 67 years). The majority presented with gastrointestinal involvement and had emergency surgery, and 40% had stage IV disease. The tumors were morphologically classified into two groups: typical (58%) and atypical (i.e., non-monomorphic or with necrosis, angiotropism or starry-sky pattern) (42%), sharing a homogeneous immunophenotypic profile (CD3+ [98%] CD4- [94%] CD5- [97%] CD7+ [97%] CD8+ [90%] CD56+ [86%] CD103+ [80%] cytotoxic marker+ [98%]) with more frequent expression of TCRgd (50%) than TCRab (32%). MYC expression (30% of cases) partly reflecting MYC gene locus alterations, correlated with non-monomorphic cytology. Almost all cases (97%) harbored deleterious mutation(s) and/or deletion of the SETD2 gene and 90% had defective H3K36 trimethylation. Other frequently mutated genes were STAT5B (57%), JAK3 (50%), TP53 (35%), JAK1 (12.5%), BCOR and ATM (11%). Both TP53 mutations and MYC expression correlated with atypical morphology. The median overall survival (OS) of 63 patients (43/63 only received chemotherapy after initial surgery) was 7.8 months. Multivariate analysis found a strong negative impact on outcome of MYC expression, TP53 mutation, STAT5B mutation and poor performance status while aberrant B-cell marker expression (20% of cases) correlated with better survival. In conclusion, MEITL is an aggressive disease with resistance to conventional therapy, predominantly characterized by driver gene alterations deregulating histone methylation and JAK/STAT signaling and encompasses genetic and morphologic variants associated with very high clinical risk.
Collapse
Affiliation(s)
- Luis Veloza
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland,LV and DC contributed equally as co-first authors
| | - Doriane Cavalieri
- Department of Hematology, University Hospital of Clermont-Ferrand, EA7453 CIC1405, Université Clermont Auvergne, Clermont-Ferrand, France,LV and DC contributed equally as co-first authors
| | - Edoardo Missiaglia
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Albane Ledoux-Pilon
- Department of Pathology, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Bettina Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Bruno Pereira
- Clinical Research Direction, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Christophe Bonnet
- Department of Hematology, University Hospital Sart Tilman, Liège, Belgium
| | - Elsa Poullot
- AP-HP, Henri Mondor Hospital, Pathology Department, Créteil, France
| | | | - Romain Dubois
- Department of Pathology, University Hospital of Lille, Lille, France
| | | | | | - Roland De Wind
- Department of Pathology, Institute Jules Bordet, Bruxelles, Belgique
| | - Fanny Drieux
- Service of Anatomical and Cytological Pathology, Center Henri Becquerel, Rouen, France
| | - Juliette Fontaine
- Multisite Pathology Institute, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Marie Parrens
- Department of Pathology, CHU de Bordeaux, University of Bordeaux, Bordeaux, France
| | - Jeremy Sandrini
- Department of Pathology, Le Mans Hospital Center, Le Mans, France
| | - Virginie Fataccioli
- AP-HP, Henri Mondor Hospital, Pathology Department, Créteil, France,University Paris Est Créteil, INSERM, IMRB, Créteil, France
| | - Marie-Hélène Delfau-Larue
- University Paris Est Créteil, INSERM, IMRB, Créteil, France,Department of Immunobiology and INSERM U955, Henri Mondor University Hospital, Créteil, France
| | - Adrien Daniel
- Department of Hematology, University Hospital of Lille, Lille, France
| | - Faustine Lhomme
- Department of Hematology, University Hospital of Rennes, Hospital Pontchaillou, Rennes, France
| | | | - François Lemonnier
- University Paris Est Créteil, INSERM, IMRB, Créteil, France,AP-HP, Henri Mondor Hospital, Lymphoid Malignancies Unit, Créteil, France
| | - Anne Cairoli
- Service of Hematology, Department of Oncology, Lausanne University, Hospital and Lausanne University, Lausanne, Switzerland
| | - Pierre Morel
- Department of Hematology, Hospital of Lens, Lens and Department of Hematology, University Hospital of Amiens, Amiens, France
| | - Sylvie Glaisner
- Department of Hematology, Institute Curie, Hospital Rene Huguenin, Saint-Cloud, France
| | - Bertrand Joly
- Department of Hematology, Sud-Francilien Hospital Center, Corbeil-Essonnes, France
| | | | - Kamel Laribi
- Department of Hematology, Hospital Center Le Mans, Le Mans, France
| | - Emmanuel Bachy
- Department of Hematology, Center Hospitalier Lyon Sud and INSERM U1111, Pierre Bénite, France
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - David Vallois
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Philippe Gaulard
- AP-HP, Henri Mondor Hospital, Pathology Department, Créteil, France,University Paris Est Créteil, INSERM, IMRB, Créteil, France,PG, OT and LdL contributed equally as co-senior authors
| | - Olivier Tournilhac
- Department of Hematology, University Hospital of Clermont-Ferrand, EA7453 CIC1405, Université Clermont Auvergne, Clermont-Ferrand, France,PG, OT and LdL contributed equally as co-senior authors
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland,PG, OT and LdL contributed equally as co-senior authors
| |
Collapse
|
23
|
In vivo impact of JAK3 A573V mutation revealed using zebrafish. Cell Mol Life Sci 2022; 79:322. [PMID: 35622134 PMCID: PMC9142468 DOI: 10.1007/s00018-022-04361-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/19/2022] [Accepted: 05/09/2022] [Indexed: 12/16/2022]
Abstract
Background Janus kinase 3 (JAK3) acts downstream of the interleukin-2 (IL-2) receptor family to play a pivotal role in the regulation of lymphoid cell development. Activating JAK3 mutations are associated with a number of lymphoid and other malignancies, with mutations within the regulatory pseudokinase domain common. Methods The pseudokinase domain mutations A572V and A573V were separately introduced into the highly conserved zebrafish Jak3 and transiently expressed in cell lines and zebrafish embryos to examine their activity and impact on early T cells. Genome editing was subsequently used to introduce the A573V mutation into the zebrafish genome to study the effects of JAK3 activation on lymphoid cells in a physiologically relevant context throughout the life-course. Results Zebrafish Jak3 A573V produced the strongest activation of downstream STAT5 in vitro and elicited a significant increase in T cells in zebrafish embryos. Zebrafish carrying just a single copy of the Jak3 A573V allele displayed elevated embryonic T cells, which continued into adulthood. Hematopoietic precursors and NK cells were also increased, but not B cells. The lymphoproliferative effects of Jak3 A573V in embryos was shown to be dependent on zebrafish IL-2Rγc, JAK1 and STAT5B equivalents, and could be suppressed with the JAK3 inhibitor Tofacitinib. Conclusions This study demonstrates that a single JAK3 A573V allele expressed from the endogenous locus was able to enhance lymphopoiesis throughout the life-course, which was mediated via an IL-2Rγc/JAK1/JAK3/STAT5 signaling pathway and was sensitive to Tofacitinib. This extends our understanding of oncogenic JAK3 mutations and creates a novel model to underpin further translational investigations. Supplementary Information The online version contains supplementary material available at 10.1007/s00018-022-04361-8.
Collapse
|
24
|
Song X, Chang S, Seminario-Vidal L, de Mingo Pulido A, Tordesillas L, Song X, Reed RA, Harkins A, Whiddon S, Nguyen JV, Segura CM, Zhang C, Yoder S, Sayegh Z, Zhao Y, Messina JL, Harro CM, Zhang X, Conejo-Garcia JR, Berglund A, Sokol L, Zhang J, Rodriguez PC, Mulé JJ, Futreal AP, Tsai KY, Chen PL. Genomic and Single-Cell Landscape Reveals Novel Drivers and Therapeutic Vulnerabilities of Transformed Cutaneous T-cell Lymphoma. Cancer Discov 2022; 12:1294-1313. [PMID: 35247891 PMCID: PMC9148441 DOI: 10.1158/2159-8290.cd-21-1207] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/10/2022] [Accepted: 02/11/2022] [Indexed: 11/16/2022]
Abstract
ABSTRACT Cutaneous T-cell lymphoma (CTCL) is a rare cancer of skin-homing T cells. A subgroup of patients develops large cell transformation with rapid progression to an aggressive lymphoma. Here, we investigated the transformed CTCL (tCTCL) tumor ecosystem using integrative multiomics spanning whole-exome sequencing (WES), single-cell RNA sequencing, and immune profiling in a unique cohort of 56 patients. WES of 70 skin biopsies showed high tumor mutation burden, UV signatures that are prognostic for survival, exome-based driver events, and most recurrently mutated pathways in tCTCL. Single-cell profiling of 16 tCTCL skin biopsies identified a core oncogenic program with metabolic reprogramming toward oxidative phosphorylation (OXPHOS), cellular plasticity, upregulation of MYC and E2F activities, and downregulation of MHC I suggestive of immune escape. Pharmacologic perturbation using OXPHOS and MYC inhibitors demonstrated potent antitumor activities, whereas immune profiling provided in situ evidence of intercellular communications between malignant T cells expressing macrophage migration inhibitory factor and macrophages and B cells expressing CD74. SIGNIFICANCE Our study contributes a key resource to the community with the largest collection of tCTCL biopsies that are difficult to obtain. The multiomics data herein provide the first comprehensive compendium of genomic alterations in tCTCL and identify potential prognostic signatures and novel therapeutic targets for an incurable T-cell lymphoma. This article is highlighted in the In This Issue feature, p. 1171.
Collapse
Affiliation(s)
- Xiaofei Song
- Department of Genomic Medicine, The UT MD Anderson Cancer Center, Houston, TX, USA
| | - Shiun Chang
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Lucia Seminario-Vidal
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Alvaro de Mingo Pulido
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Leticia Tordesillas
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Xingzhi Song
- Department of Genomic Medicine, The UT MD Anderson Cancer Center, Houston, TX, USA
| | - Rhianna A. Reed
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Andrea Harkins
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Shannen Whiddon
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jonathan V. Nguyen
- Advanced Analytical and Digital Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Carlos Moran Segura
- Advanced Analytical and Digital Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Chaomei Zhang
- Molecular Genomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sean Yoder
- Molecular Genomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Zena Sayegh
- Tissue Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yun Zhao
- Department of Biopharma Services, Admera Health, Holmdel, NJ, USA
| | - Jane L. Messina
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Carly M. Harro
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Xiaohui Zhang
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - José R. Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Anders Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Lubomir Sokol
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The UT MD Anderson Cancer Center, Houston, TX, USA
| | - Paulo C. Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - James J. Mulé
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Andrew P. Futreal
- Department of Genomic Medicine, The UT MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth Y. Tsai
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Pei-Ling Chen
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
25
|
Isabelle C, Boles A, Chakravarti N, Porcu P, Brammer J, Mishra A. Cytokines in the Pathogenesis of Large Granular Lymphocytic Leukemia. Front Oncol 2022; 12:849917. [PMID: 35359386 PMCID: PMC8960188 DOI: 10.3389/fonc.2022.849917] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 12/25/2022] Open
Abstract
Large granular lymphocytic leukemia (LGLL) is a lymphoproliferative disorder of older adults characterized by the clonal expansion of cytotoxic T/natural killer cells due to constitutive pro-survival signaling. In recent years, it has become clear that cytokines and their receptors are aberrantly expressed in LGLL cells. The exact initiation process of LGLL is unknown, although several cytokine-driven mechanisms have emerged. Elevated levels of several cytokines, including interleukin-15 (IL-15) and platelet-derived growth factor (PDGF), have been described in LGLL patients. Evidence from humans and animal models has shown that cytokines may also contribute to the co-occurrence of a wide range of autoimmune diseases seen in patients with LGLL. The goal of this review is to provide a comprehensive analysis of the link between cytokines and pro-survival signaling in LGLL and to discuss the various strategies and research approaches that are being utilized to study this link. This review will also highlight the importance of cytokine-targeted therapeutics in the treatment of LGLL.
Collapse
Affiliation(s)
- Colleen Isabelle
- Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Amy Boles
- Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Nitin Chakravarti
- Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Pierluigi Porcu
- Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Jonathan Brammer
- Division of Hematology, The Ohio State University, Columbus, OH, United States
| | - Anjali Mishra
- Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Philadelphia, PA, United States
- *Correspondence: Anjali Mishra,
| |
Collapse
|
26
|
Erdogan F, Qadree AK, Radu TB, Orlova A, de Araujo ED, Israelian J, Valent P, Mustjoki SM, Herling M, Moriggl R, Gunning PT. Structural and mutational analysis of member-specific STAT functions. Biochim Biophys Acta Gen Subj 2022; 1866:130058. [PMID: 34774983 DOI: 10.1016/j.bbagen.2021.130058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND The STAT family of transcription factors control gene expression in response to signals from various stimulus. They display functions in diseases ranging from autoimmunity and chronic inflammatory disease to cancer and infectious disease. SCOPE OF REVIEW This work uses an approach informed by structural data to explore how domain-specific structural variations, post-translational modifications, and the cancer genome mutational landscape dictate STAT member-specific activities. MAJOR CONCLUSIONS We illustrated the structure-function relationship of STAT proteins and highlighted their effect on member-specific activity. We correlated disease-linked STAT mutations to the structure and cancer genome mutational landscape and proposed rational drug targeting approaches of oncogenic STAT pathway addiction. GENERAL SIGNIFICANCE Hyper-activated STATs and their variants are associated with multiple diseases and are considered high value oncology targets. A full understanding of the molecular basis of member-specific STAT-mediated signaling and the strategies to selectively target them requires examination of the difference in their structures and sequences.
Collapse
Affiliation(s)
- Fettah Erdogan
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Canada
| | - Abdul K Qadree
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Canada
| | - Tudor B Radu
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Canada
| | - Anna Orlova
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria
| | - Elvin D de Araujo
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada
| | - Johan Israelian
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Canada
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Satu M Mustjoki
- Hematology Research Unit, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland; Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Marco Herling
- Department of Hematology, Cellular Therapy, and Hemostaseology, University of Leipzig, Leipzig, Germany
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria
| | - Patrick T Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359 Mississauga Rd N., Mississauga, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Canada.
| |
Collapse
|
27
|
Schönefeldt S, Wais T, Herling M, Mustjoki S, Bekiaris V, Moriggl R, Neubauer HA. The Diverse Roles of γδ T Cells in Cancer: From Rapid Immunity to Aggressive Lymphoma. Cancers (Basel) 2021; 13:6212. [PMID: 34944832 PMCID: PMC8699114 DOI: 10.3390/cancers13246212] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022] Open
Abstract
γδ T cells are unique players in shaping immune responses, lying at the intersection between innate and adaptive immunity. Unlike conventional αβ T cells, γδ T cells largely populate non-lymphoid peripheral tissues, demonstrating tissue specificity, and they respond to ligands in an MHC-independent manner. γδ T cells display rapid activation and effector functions, with a capacity for cytotoxic anti-tumour responses and production of inflammatory cytokines such as IFN-γ or IL-17. Their rapid cytotoxic nature makes them attractive cells for use in anti-cancer immunotherapies. However, upon transformation, γδ T cells can give rise to highly aggressive lymphomas. These rare malignancies often display poor patient survival, and no curative therapies exist. In this review, we discuss the diverse roles of γδ T cells in immune surveillance and response, with a particular focus on cancer immunity. We summarise the intriguing dichotomy between pro- and anti-tumour functions of γδ T cells in solid and haematological cancers, highlighting the key subsets involved. Finally, we discuss potential drivers of γδ T-cell transformation, summarising the main γδ T-cell lymphoma/leukaemia entities, their clinical features, recent advances in mapping their molecular and genomic landscapes, current treatment strategies and potential future targeting options.
Collapse
Affiliation(s)
- Susann Schönefeldt
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (S.S.); (T.W.); (R.M.)
| | - Tamara Wais
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (S.S.); (T.W.); (R.M.)
| | - Marco Herling
- Department of Hematology, Cellular Therapy and Hemostaseology, University of Leipzig, 04103 Leipzig, Germany;
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland;
- iCAN Digital Precision Cancer Medicine Flagship, 00014 Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland
| | - Vasileios Bekiaris
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark;
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (S.S.); (T.W.); (R.M.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (S.S.); (T.W.); (R.M.)
| |
Collapse
|
28
|
Sreedharanunni S, Jamwal M, Balakrishnan A, Aravindan AV, Sharma R, Singh N, Rajpal S, Singla S, Khadwal AR, Ahluwalia J, Malhotra P, Das R. Chronic eosinophilic leukemia with recurrent STAT5B N642H mutation-An entity with features of myelodysplastic syndrome/ myeloproliferative neoplasm overlap. Leuk Res 2021; 112:106753. [PMID: 34856508 DOI: 10.1016/j.leukres.2021.106753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Sreejesh Sreedharanunni
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Manu Jamwal
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anand Balakrishnan
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arun Vijayalakshmi Aravindan
- Adult Clinical Hematology Unit, Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ritika Sharma
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Namrata Singh
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sweta Rajpal
- Department of Hematopathology, ACTREC, Navi Mumbai, India
| | - Shelly Singla
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Rani Khadwal
- Adult Clinical Hematology Unit, Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jasmina Ahluwalia
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Malhotra
- Adult Clinical Hematology Unit, Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Reena Das
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
29
|
Liu Z, Qing P, Zhao Y, Liu Y, Marion TN. Combined Mutation of the GATA2 Gene and STAT5B Gene in a Patient with Hypogammaglobulinemia and Autoimmunity. TOHOKU J EXP MED 2021; 255:143-146. [PMID: 34670919 DOI: 10.1620/tjem.255.143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Antibody deficiency is a type of primary immunodeficiency that often manifests as primary hypogammaglobulinemia, with or without repeated infections. Although primary immunodeficiency appears to be contrary to autoimmunity, they usually occur simultaneously, and the specific pathogenesis remains unknown. We herein describe an adult patient with autoimmune manifestations and recurrent infections. The case was characterized by a sustained decrease in serum immunoglobulin A, accompanied by decreased T lymphocytes, B lymphocytes, monocytes, and platelets in the peripheral blood and the presence of antinuclear and anti-SSA antibodies. Whole-exome sequencing for the patient revealed two spontaneous mutations in GATA2 (c.1084C>T) and STAT5B (c.1924A>C). This case report provides evidence that mutations in the GATA2 and STAT5B genes may be pathogenic in primary immunodeficiency and provides genetic evidence for the possible pathogenesis of primary immunodeficiency with autoimmune symptoms. However, further studies are needed to confirm the causal relationship.
Collapse
Affiliation(s)
- Zhihui Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University
| | - Pingying Qing
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University
| | - Tony N Marion
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University.,Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center
| |
Collapse
|
30
|
Kieslinger M, Swoboda A, Kramer N, Freund P, Pratscher B, Neubauer HA, Steinborn R, Wolfesberger B, Fuchs-Baumgartinger A, Moriggl R, Burgener IA. A Recurrent STAT5BN642H Driver Mutation in Feline Alimentary T Cell Lymphoma. Cancers (Basel) 2021; 13:5238. [PMID: 34680385 PMCID: PMC8534107 DOI: 10.3390/cancers13205238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/05/2021] [Indexed: 11/18/2022] Open
Abstract
Alimentary lymphomas arising from T cells are rare and aggressive malignancies in humans. In comparison, they represent the most common anatomical form of lymphoma in cats. Due to the low prevalence in humans, the underlying pathomechanism for these diseases is poorly characterised, limiting experimental analysis and therapeutic exploration. To date, activating mutations of the JAK/STAT core cancer pathway and particularly the STAT5B oncoprotein have been identified in human enteropathy-associated T cell lymphoma. Here, we describe a high homology of human and feline STAT3 and STAT5B proteins and strong conservation at the genomic level. Analysis of 42 samples of feline T cell alimentary lymphoma reveals broad activation of STAT3 and STAT5B. Screening for known activating mutations in STAT3 or STAT5B identifies the presence of the STAT5BN642H driver mutation in feline enteropathy-associated T cell lymphoma in 7 out of 42 (16.67%) samples in total. Regarding lymphoma subtypes, the majority of mutations with 5 out of 17 (29.41%) cases were found in feline enteropathy-associated lymphoma type II (EATL II). This identification of an oncogenic STAT5B driver mutation in felines recapitulates the genetic situation in the corresponding human disease, thereby establishing the cat as a potential new model for a rare and incurable human T cell disease.
Collapse
Affiliation(s)
- Matthias Kieslinger
- Department for Companion Animals and Horses, Division of Small Animal Internal Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (A.S.); (N.K.); (P.F.); (B.P.); (B.W.); (I.A.B.)
| | - Alexander Swoboda
- Department for Companion Animals and Horses, Division of Small Animal Internal Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (A.S.); (N.K.); (P.F.); (B.P.); (B.W.); (I.A.B.)
| | - Nina Kramer
- Department for Companion Animals and Horses, Division of Small Animal Internal Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (A.S.); (N.K.); (P.F.); (B.P.); (B.W.); (I.A.B.)
| | - Patricia Freund
- Department for Companion Animals and Horses, Division of Small Animal Internal Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (A.S.); (N.K.); (P.F.); (B.P.); (B.W.); (I.A.B.)
| | - Barbara Pratscher
- Department for Companion Animals and Horses, Division of Small Animal Internal Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (A.S.); (N.K.); (P.F.); (B.P.); (B.W.); (I.A.B.)
| | - Heidi A. Neubauer
- Department of Biomedical Sciences, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (H.A.N.); (R.M.)
| | - Ralf Steinborn
- Genomics Core Facility, VetCore, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Birgitt Wolfesberger
- Department for Companion Animals and Horses, Division of Small Animal Internal Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (A.S.); (N.K.); (P.F.); (B.P.); (B.W.); (I.A.B.)
| | - Andrea Fuchs-Baumgartinger
- Department of Pathobiology, Institute of Pathology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Richard Moriggl
- Department of Biomedical Sciences, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (H.A.N.); (R.M.)
| | - Iwan A. Burgener
- Department for Companion Animals and Horses, Division of Small Animal Internal Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (A.S.); (N.K.); (P.F.); (B.P.); (B.W.); (I.A.B.)
| |
Collapse
|
31
|
A STAT5B-CD9 axis determines self-renewal in hematopoietic and leukemic stem cells. Blood 2021; 138:2347-2359. [PMID: 34320169 DOI: 10.1182/blood.2021010980] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 07/21/2021] [Indexed: 11/20/2022] Open
Abstract
The transcription factors STAT5A and STAT5B are critical in hematopoiesis and leukemia. They are widely believed to have redundant functions but we describe a unique role for STAT5B in driving the self-renewal of hematopoietic and leukemic stem cells (HSCs/LSCs). We find STAT5B to be specifically activated in HSCs and LSCs, where it induces many genes associated with quiescence and self-renewal, including the surface marker CD9. Levels of CD9 represent a prognostic marker for patients with STAT5-driven leukemia and our findings suggest that anti-CD9 antibodies may be useful in their treatment to target and eliminate LSCs. We show that it is vital to consider STAT5A and STAT5B as distinct entities in normal and malignant hematopoiesis.
Collapse
|
32
|
STAT5 as a Key Protein of Erythropoietin Signalization. Int J Mol Sci 2021; 22:ijms22137109. [PMID: 34281163 PMCID: PMC8268974 DOI: 10.3390/ijms22137109] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 12/14/2022] Open
Abstract
Erythropoietin (EPO) acts on multiple tissues through its receptor EPOR, a member of a cytokine class I receptor superfamily with pleiotropic effects. The interaction of EPO and EPOR triggers the activation of several signaling pathways that induce erythropoiesis, including JAK2/STAT5, PI3K/AKT, and MAPK. The canonical EPOR/JAK2/STAT5 pathway is a known regulator of differentiation, proliferation, and cell survival of erythroid progenitors. In addition, its role in the protection of other cells, including cancer cells, is under intense investigation. The involvement of EPOR/JAK2/STAT5 in other processes such as mRNA splicing, cytoskeleton reorganization, and cell metabolism has been recently described. The transcriptomics, proteomics, and epigenetic studies reviewed in this article provide a detailed understanding of EPO signalization. Advances in this area of research may be useful for improving the efficacy of EPO therapy in hematologic disorders, as well as in cancer treatment.
Collapse
|
33
|
Untwining Anti-Tumor and Immunosuppressive Effects of JAK Inhibitors-A Strategy for Hematological Malignancies? Cancers (Basel) 2021; 13:cancers13112611. [PMID: 34073410 PMCID: PMC8197909 DOI: 10.3390/cancers13112611] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/18/2021] [Accepted: 05/22/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is aberrantly activated in many malignancies. Inhibition of this pathway via JAK inhibitors (JAKinibs) is therefore an attractive therapeutic strategy underlined by Ruxolitinib (JAK1/2 inhibitor) being approved for the treatment of myeloproliferative neoplasms. As a consequence of the crucial role of the JAK-STAT pathway in the regulation of immune responses, inhibition of JAKs suppresses the immune system. This review article provides a thorough overview of the current knowledge on JAKinibs’ effects on immune cells in the context of hematological malignancies. We also discuss the potential use of JAKinibs for the treatment of diseases in which lymphocytes are the source of the malignancy. Abstract The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway propagates signals from a variety of cytokines, contributing to cellular responses in health and disease. Gain of function mutations in JAKs or STATs are associated with malignancies, with JAK2V617F being the main driver mutation in myeloproliferative neoplasms (MPN). Therefore, inhibition of this pathway is an attractive therapeutic strategy for different types of cancer. Numerous JAK inhibitors (JAKinibs) have entered clinical trials, including the JAK1/2 inhibitor Ruxolitinib approved for the treatment of MPN. Importantly, loss of function mutations in JAK-STAT members are a cause of immune suppression or deficiencies. MPN patients undergoing Ruxolitinib treatment are more susceptible to infections and secondary malignancies. This highlights the suppressive effects of JAKinibs on immune responses, which renders them successful in the treatment of autoimmune diseases but potentially detrimental for cancer patients. Here, we review the current knowledge on the effects of JAKinibs on immune cells in the context of hematological malignancies. Furthermore, we discuss the potential use of JAKinibs for the treatment of diseases in which lymphocytes are the source of malignancies. In summary, this review underlines the necessity of a robust immune profiling to provide the best benefit for JAKinib-treated patients.
Collapse
|
34
|
Deregulation of the Interleukin-7 Signaling Pathway in Lymphoid Malignancies. Pharmaceuticals (Basel) 2021; 14:ph14050443. [PMID: 34066732 PMCID: PMC8151260 DOI: 10.3390/ph14050443] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/21/2022] Open
Abstract
The cytokine interleukin-7 (IL-7) and its receptor are critical for lymphoid cell development. The loss of IL-7 signaling causes severe combined immunodeficiency, whereas gain-of-function alterations in the pathway contribute to malignant transformation of lymphocytes. Binding of IL-7 to the IL-7 receptor results in the activation of the JAK-STAT, PI3K-AKT and Ras-MAPK pathways, each contributing to survival, cell cycle progression, proliferation and differentiation. Here, we discuss the role of deregulated IL-7 signaling in lymphoid malignancies of B- and T-cell origin. Especially in T-cell leukemia, more specifically in T-cell acute lymphoblastic leukemia and T-cell prolymphocytic leukemia, a high frequency of mutations in components of the IL-7 signaling pathway are found, including alterations in IL7R, IL2RG, JAK1, JAK3, STAT5B, PTPN2, PTPRC and DNM2 genes.
Collapse
|
35
|
Polomski M, Brachet-Botineau M, Juen L, Viaud-Massuard MC, Gouilleux F, Prié G. Inhibitors Targeting STAT5 Signaling in Myeloid Leukemias: New Tetrahydroquinoline Derivatives with Improved Antileukemic Potential. ChemMedChem 2021; 16:1034-1046. [PMID: 33275308 DOI: 10.1002/cmdc.202000841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Indexed: 01/23/2023]
Abstract
Signal transducers and activators of transcription 5A and 5B (STAT5A and STAT5B) are two closely related STAT family members that are crucial downstream effectors of tyrosine kinase oncoproteins such as FLT3-ITD in acute myeloid leukemia (AML) and BCR-ABL in chronic myeloid leukemia (CML). We recently developed and reported the synthesis of a first molecule called 17 f that selectively inhibits STAT5 signaling in myeloid leukemia cells and overcomes their resistance to chemotherapeutic agents. To improve the antileukemic effect of 17 f, we synthesized ten analogs of this molecule and analyzed their impact on cell growth, survival, chemoresistance and STAT5 signaling. Two compounds, 7 a and 7 a', were identified as having similar or higher antileukemic effects in various AML and CML cell lines. Both molecules were found to be more effective than 17 f at inhibiting STAT5 activity/expression and suppressing the chemoresistance of CML.
Collapse
Affiliation(s)
- Marion Polomski
- Equipe IMT "Innovation Moléculaire et Thérapeutique" - GICC EA7501, Université de Tours-Labex SYNORG, Faculté de Pharmacie, 31 av. Monge, 37200, Tours, France
| | - Marie Brachet-Botineau
- Equipe LNOx "Niche Leucémique & Métabolisme Oxydatif" - GICC ERL 7001 CNRS, Université de Tours, Faculté de Médecine, Bâtiment Dutrochet, 10bis bvd Tonnellé, 37032, Tours, France
| | - Ludovic Juen
- Equipe IMT "Innovation Moléculaire et Thérapeutique" - GICC EA7501, Université de Tours-Labex SYNORG, Faculté de Pharmacie, 31 av. Monge, 37200, Tours, France
| | - Marie-Claude Viaud-Massuard
- Equipe IMT "Innovation Moléculaire et Thérapeutique" - GICC EA7501, Université de Tours-Labex SYNORG, Faculté de Pharmacie, 31 av. Monge, 37200, Tours, France
| | - Fabrice Gouilleux
- Equipe LNOx "Niche Leucémique & Métabolisme Oxydatif" - GICC ERL 7001 CNRS, Université de Tours, Faculté de Médecine, Bâtiment Dutrochet, 10bis bvd Tonnellé, 37032, Tours, France
| | - Gildas Prié
- Equipe IMT "Innovation Moléculaire et Thérapeutique" - GICC EA7501, Université de Tours-Labex SYNORG, Faculté de Pharmacie, 31 av. Monge, 37200, Tours, France
| |
Collapse
|
36
|
Shalini, Kumar V. Have molecular hybrids delivered effective anti-cancer treatments and what should future drug discovery focus on? Expert Opin Drug Discov 2020; 16:335-363. [PMID: 33305635 DOI: 10.1080/17460441.2021.1850686] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Cancer continues to be a big threat and its treatment is a huge challenge among the medical fraternity. Conventional anti-cancer agents are losing their efficiency which highlights the need to introduce new anti-cancer entities for treating this complex disease. A hybrid molecule has a tendency to act through varied modes of action on multiple targets at a given time. Thus, there is the significant scope with hybrid compounds to tackle the existing limitations of cancer chemotherapy. AREA COVERED This perspective describes the most significant hybrids that spring hope in the field of cancer chemotherapy. Several hybrids with anti-proliferative/anti-tumor properties currently approved or in clinical development are outlined, along with a description of their mechanism of action and identified drug targets. EXPERT OPINION The success of molecular hybridization in cancer chemotherapy is quite evident by the number of molecules entering into clinical trials and/or have entered the drug market over the past decade. Indeed, the recent advancements and co-ordinations in the interface between chemistry, biology, and pharmacology will help further the advancement of hybrid chemotherapeutics in the future.List of abbreviations: Deoxyribonucleic acid, DNA; national cancer institute, NCI; peripheral blood mononuclear cells, PBMC; food and drug administration, FDA; histone deacetylase, HDAC; epidermal growth factor receptor, EGFR; vascular endothelial growth factor receptor, VEGFR; suberoylanilide hydroxamic acid, SAHA; farnesyltransferase inhibitor, FTI; adenosine triphosphate, ATP; Tamoxifen, TAM; selective estrogen receptor modulator, SERM; structure activity relationship, SAR; estrogen receptor, ER; lethal dose, LD; half maximal growth inhibitory concentration, GI50; half maximal inhibitory concentration, IC50.
Collapse
Affiliation(s)
- Shalini
- Department of Chemistry, Guru Nanak Dev University, Amritsar-India
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar-India
| |
Collapse
|
37
|
Ding ZC, Shi H, Aboelella NS, Fesenkova K, Park EJ, Liu Z, Pei L, Li J, McIndoe RA, Xu H, Piazza GA, Blazar BR, Munn DH, Zhou G. Persistent STAT5 activation reprograms the epigenetic landscape in CD4 + T cells to drive polyfunctionality and antitumor immunity. Sci Immunol 2020; 5:eaba5962. [PMID: 33127608 PMCID: PMC8265158 DOI: 10.1126/sciimmunol.aba5962] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 06/21/2020] [Accepted: 10/08/2020] [Indexed: 01/05/2023]
Abstract
The presence of polyfunctional CD4+ T cells is often associated with favorable antitumor immunity. We report here that persistent activation of signal transducer and activator of transcription 5 (STAT5) in tumor-specific CD4+ T cells drives the development of polyfunctional T cells. We showed that ectopic expression of a constitutively active form of murine STAT5A (CASTAT5) enabled tumor-specific CD4+ T cells to undergo robust expansion, infiltrate tumors vigorously, and elicit antitumor CD8+ T cell responses in a CD4+ T cell adoptive transfer model system. Integrated epigenomic and transcriptomic analysis revealed that CASTAT5 induced genome-wide chromatin remodeling in CD4+ T cells and established a distinct epigenetic and transcriptional landscape. Single-cell RNA sequencing analysis further identified a subset of CASTAT5-transduced CD4+ T cells with a molecular signature indicative of progenitor polyfunctional T cells. The therapeutic significance of CASTAT5 came from our finding that adoptive transfer of T cells engineered to coexpress CD19-targeting chimeric antigen receptor (CAR) and CASTAT5 gave rise to polyfunctional CD4+ CAR T cells in a mouse B cell lymphoma model. The optimal therapeutic outcome was obtained when both CD4+ and CD8+ CAR T cells were transduced with CASTAT5, indicating that CASTAT5 facilitates productive CD4 help to CD8+ T cells. Furthermore, we provide evidence that CASTAT5 is functional in primary human CD4+ T cells, underscoring its potential clinical relevance. Our results implicate STAT5 as a valid candidate for T cell engineering to generate polyfunctional, exhaustion-resistant, and tumor-tropic antitumor CD4+ T cells to potentiate adoptive T cell therapy for cancer.
Collapse
Affiliation(s)
- Zhi-Chun Ding
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| | - Huidong Shi
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA.
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Nada S Aboelella
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Kateryna Fesenkova
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Eun-Jeong Park
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Zhuoqi Liu
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Lirong Pei
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jiaqi Li
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Richard A McIndoe
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Hongyan Xu
- Division of Biostatistics and Data Science, Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Gary A Piazza
- University of South Alabama Mitchell Cancer Institute, Mobile, AL, USA
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - David H Munn
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Gang Zhou
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, USA.
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
38
|
Kadekar D, Agerholm R, Rizk J, Neubauer HA, Suske T, Maurer B, Viñals MT, Comelli EM, Taibi A, Moriggl R, Bekiaris V. The neonatal microenvironment programs innate γδ T cells through the transcription factor STAT5. J Clin Invest 2020; 130:2496-2508. [PMID: 32281944 PMCID: PMC7190909 DOI: 10.1172/jci131241] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 01/29/2020] [Indexed: 01/18/2023] Open
Abstract
IL-17-producing RORγt+ γδ T cells (γδT17 cells) are innate lymphocytes that participate in type 3 immune responses during infection and inflammation. Herein, we show that γδT17 cells rapidly proliferate within neonatal lymph nodes and gut, where, upon entry, they upregulate T-bet and coexpress IL-17, IL-22, and IFN-γ in a STAT3- and retinoic acid-dependent manner. Neonatal expansion was halted in mice conditionally deficient in STAT5, and its loss resulted in γδT17 cell depletion from all adult organs. Hyperactive STAT5 mutant mice showed that the STAT5A homolog had a dominant role over STAT5B in promoting γδT17 cell expansion and downregulating gut-associated T-bet. In contrast, STAT5B preferentially expanded IFN-γ-producing γδ populations, implying a previously unknown differential role of STAT5 gene products in lymphocyte lineage regulation. Importantly, mice lacking γδT17 cells as a result of STAT5 deficiency displayed a profound resistance to experimental autoimmune encephalomyelitis. Our data identify that the neonatal microenvironment in combination with STAT5 is critical for post-thymic γδT17 development and tissue-specific imprinting, which is essential for infection and autoimmunity.
Collapse
Affiliation(s)
- Darshana Kadekar
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Rasmus Agerholm
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - John Rizk
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Tobias Suske
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Barbara Maurer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Elena M. Comelli
- Department of Nutritional Sciences and
- Department of Nutritional Sciences and Joannah and Brian Lawson Centre for Child Nutrition, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Vasileios Bekiaris
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
39
|
Teramo A, Barilà G, Calabretto G, Vicenzetto C, Gasparini VR, Semenzato G, Zambello R. Insights Into Genetic Landscape of Large Granular Lymphocyte Leukemia. Front Oncol 2020; 10:152. [PMID: 32133291 PMCID: PMC7040228 DOI: 10.3389/fonc.2020.00152] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/28/2020] [Indexed: 01/29/2023] Open
Abstract
Large granular lymphocyte leukemia (LGLL) is a chronic proliferation of clonal cytotoxic lymphocytes, usually presenting with cytopenias and yet lacking a specific therapy. The disease is heterogeneous, including different subsets of patients distinguished by LGL immunophenotype (CD8+ Tαβ, CD4+ Tαβ, Tγδ, NK) and the clinical course of the disease (indolent/symptomatic/aggressive). Even if the etiology of LGLL remains elusive, evidence is accumulating on the genetic landscape driving and/or sustaining chronic LGL proliferations. The most common gain-of-function mutations identified in LGLL patients are on STAT3 and STAT5b genes, which have been recently recognized as clonal markers and were included in the 2017 WHO classification of the disease. A significant correlation between STAT3 mutations and symptomatic disease has been highlighted. At variance, STAT5b mutations could have a different clinical impact based on the immunophenotype of the mutated clone. In fact, they are regarded as the signature of an aggressive clinical course with a poor prognosis in CD8+ T-LGLL and aggressive NK cell leukemia, while they are devoid of negative prognostic significance in CD4+ T-LGLL and Tγδ LGLL. Knowing the specific distribution of STAT mutations helps identify the discrete mechanisms sustaining LGL proliferations in the corresponding disease subsets. Some patients equipped with wild type STAT genes are characterized by less frequent mutations in different genes, suggesting that other pathogenetic mechanisms are likely to be involved. In this review, we discuss how the LGLL mutational pattern allows a more precise and detailed tumor stratification, suggesting new parameters for better management of the disease and hopefully paving the way for a targeted clinical approach.
Collapse
Affiliation(s)
- Antonella Teramo
- Hematology and Clinical Immunology Section, Department of Medicine (DIMED), Padova University School of Medicine, Padova, Italy.,Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Gregorio Barilà
- Hematology and Clinical Immunology Section, Department of Medicine (DIMED), Padova University School of Medicine, Padova, Italy.,Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Giulia Calabretto
- Hematology and Clinical Immunology Section, Department of Medicine (DIMED), Padova University School of Medicine, Padova, Italy.,Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Cristina Vicenzetto
- Hematology and Clinical Immunology Section, Department of Medicine (DIMED), Padova University School of Medicine, Padova, Italy.,Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Vanessa Rebecca Gasparini
- Hematology and Clinical Immunology Section, Department of Medicine (DIMED), Padova University School of Medicine, Padova, Italy.,Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Gianpietro Semenzato
- Hematology and Clinical Immunology Section, Department of Medicine (DIMED), Padova University School of Medicine, Padova, Italy.,Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Renato Zambello
- Hematology and Clinical Immunology Section, Department of Medicine (DIMED), Padova University School of Medicine, Padova, Italy.,Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| |
Collapse
|
40
|
Maurer B, Nivarthi H, Wingelhofer B, Pham HTT, Schlederer M, Suske T, Grausenburger R, Schiefer AI, Prchal-Murphy M, Chen D, Winkler S, Merkel O, Kornauth C, Hofbauer M, Hochgatterer B, Hoermann G, Hoelbl-Kovacic A, Prochazkova J, Lobello C, Cumaraswamy AA, Latzka J, Kitzwögerer M, Chott A, Janikova A, Pospíšilova Š, Loizou JI, Kubicek S, Valent P, Kolbe T, Grebien F, Kenner L, Gunning PT, Kralovics R, Herling M, Müller M, Rülicke T, Sexl V, Moriggl R. High activation of STAT5A drives peripheral T-cell lymphoma and leukemia. Haematologica 2020; 105:435-447. [PMID: 31123029 PMCID: PMC7012494 DOI: 10.3324/haematol.2019.216986] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/21/2019] [Indexed: 12/11/2022] Open
Abstract
Recurrent gain-of-function mutations in the transcription factors STAT5A and much more in STAT5B were found in hematopoietic malignancies with the highest proportion in mature T- and natural killer-cell neoplasms (peripheral T-cell lymphoma, PTCL). No targeted therapy exists for these heterogeneous and often aggressive diseases. Given the shortage of models for PTCL, we mimicked graded STAT5A or STAT5B activity by expressing hyperactive Stat5a or STAT5B variants at low or high levels in the hematopoietic system of transgenic mice. Only mice with high activity levels developed a lethal disease resembling human PTCL. Neoplasia displayed massive expansion of CD8+ T cells and destructive organ infiltration. T cells were cytokine-hypersensitive with activated memory CD8+ T-lymphocyte characteristics. Histopathology and mRNA expression profiles revealed close correlation with distinct subtypes of PTCL. Pronounced STAT5 expression and activity in samples from patients with different subsets underline the relevance of JAK/STAT as a therapeutic target. JAK inhibitors or a selective STAT5 SH2 domain inhibitor induced cell death and ruxolitinib blocked T-cell neoplasia in vivo. We conclude that enhanced STAT5A or STAT5B action both drive PTCL development, defining both STAT5 molecules as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Barbara Maurer
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria.,Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Harini Nivarthi
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bettina Wingelhofer
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ha Thi Thanh Pham
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Michaela Schlederer
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Department of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Tobias Suske
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Reinhard Grausenburger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ana-Iris Schiefer
- Department of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Michaela Prchal-Murphy
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Doris Chen
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Susanne Winkler
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Olaf Merkel
- Department of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Christoph Kornauth
- Department of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | | | | | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Andrea Hoelbl-Kovacic
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Jana Prochazkova
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Cosimo Lobello
- Central European Institute of Technology (CEITEC), Center of Molecular Medicine, Masaryk University, Brno, Czech Republic
| | - Abbarna A Cumaraswamy
- Department of Chemistry, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Johanna Latzka
- Karl Landsteiner Institute of Dermatological Research, St. Poelten, Austria and Department of Dermatology and Venereology, Karl Landsteiner University for Health Sciences, St. Poelten, Austria
| | - Melitta Kitzwögerer
- Department of Clinical Pathology, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Andreas Chott
- Institute of Pathology and Microbiology, Wilheminenspital, Vienna, Austria
| | - Andrea Janikova
- Department of Internal Medicine - Hematology and Oncology, Faculty of Medicine Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Šárka Pospíšilova
- Central European Institute of Technology (CEITEC), Center of Molecular Medicine, Masaryk University, Brno, Czech Republic.,Department of Internal Medicine - Hematology and Oncology, Faculty of Medicine Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Joanna I Loizou
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology and Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - Thomas Kolbe
- Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria.,IFA-Tulln, University of Natural Resources and Applied Life Sciences, Tulln, Austria
| | - Florian Grebien
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Lukas Kenner
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Department of Clinical Pathology, Medical University of Vienna, Vienna, Austria.,Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Patrick T Gunning
- Central European Institute of Technology (CEITEC), Center of Molecular Medicine, Masaryk University, Brno, Czech Republic
| | - Robert Kralovics
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Marco Herling
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Köln-Bonn, Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria .,Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria.,Medical University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
Brachet-Botineau M, Polomski M, Neubauer HA, Juen L, Hédou D, Viaud-Massuard MC, Prié G, Gouilleux F. Pharmacological Inhibition of Oncogenic STAT3 and STAT5 Signaling in Hematopoietic Cancers. Cancers (Basel) 2020; 12:E240. [PMID: 31963765 PMCID: PMC7016966 DOI: 10.3390/cancers12010240] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
Signal Transducer and Activator of Transcription (STAT) 3 and 5 are important effectors of cellular transformation, and aberrant STAT3 and STAT5 signaling have been demonstrated in hematopoietic cancers. STAT3 and STAT5 are common targets for different tyrosine kinase oncogenes (TKOs). In addition, STAT3 and STAT5 proteins were shown to contain activating mutations in some rare but aggressive leukemias/lymphomas. Both proteins also contribute to drug resistance in hematopoietic malignancies and are now well recognized as major targets in cancer treatment. The development of inhibitors targeting STAT3 and STAT5 has been the subject of intense investigations during the last decade. This review summarizes the current knowledge of oncogenic STAT3 and STAT5 functions in hematopoietic cancers as well as advances in preclinical and clinical development of pharmacological inhibitors.
Collapse
Affiliation(s)
- Marie Brachet-Botineau
- Leukemic Niche and Oxidative metabolism (LNOx), CNRS ERL 7001, University of Tours, 37000 Tours, France;
| | - Marion Polomski
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, A-1210 Vienna, Austria;
| | - Ludovic Juen
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Damien Hédou
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Marie-Claude Viaud-Massuard
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Gildas Prié
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Fabrice Gouilleux
- Leukemic Niche and Oxidative metabolism (LNOx), CNRS ERL 7001, University of Tours, 37000 Tours, France;
| |
Collapse
|
42
|
Brachet-Botineau M, Deynoux M, Vallet N, Polomski M, Juen L, Hérault O, Mazurier F, Viaud-Massuard MC, Prié G, Gouilleux F. A Novel Inhibitor of STAT5 Signaling Overcomes Chemotherapy Resistance in Myeloid Leukemia Cells. Cancers (Basel) 2019; 11:cancers11122043. [PMID: 31861239 PMCID: PMC6966442 DOI: 10.3390/cancers11122043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/11/2019] [Accepted: 12/14/2019] [Indexed: 01/08/2023] Open
Abstract
Signal transducers and activators of transcription 5A and 5B (STAT5A and STAT5B) are crucial downstream effectors of tyrosine kinase oncogenes (TKO) such as BCR-ABL in chronic myeloid leukemia (CML) and FLT3-ITD in acute myeloid leukemia (AML). Both proteins have been shown to promote the resistance of CML cells to tyrosine kinase inhibitors (TKI) such as imatinib mesylate (IM). We recently synthesized and discovered a new inhibitor (17f) with promising antileukemic activity. 17f selectively inhibits STAT5 signaling in CML and AML cells by interfering with the phosphorylation and transcriptional activity of these proteins. In this study, the effects of 17f were evaluated on CML and AML cell lines that respectively acquired resistance to IM and cytarabine (Ara-C), a conventional therapeutic agent used in AML treatment. We showed that 17f strongly inhibits the growth and survival of resistant CML and AML cells when associated with IM or Ara-C. We also obtained evidence that 17f inhibits STAT5B but not STAT5A protein expression in resistant CML and AML cells. Furthermore, we demonstrated that 17f also targets oncogenic STAT5B N642H mutant in transformed hematopoietic cells.
Collapse
Affiliation(s)
- Marie Brachet-Botineau
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
| | - Margaux Deynoux
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
| | - Nicolas Vallet
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
- Service d’Hématologie et Thérapie Cellulaire, CHRU de Tours, 37000 Tours, France
| | - Marion Polomski
- IMT, GICC, EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (M.-C.V.-M.); (G.P.)
| | - Ludovic Juen
- IMT, GICC, EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (M.-C.V.-M.); (G.P.)
| | - Olivier Hérault
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
- Service d’Hematologie Biologique, CHRU de Tours, 37000 Tours, France
| | - Frédéric Mazurier
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
| | | | - Gildas Prié
- IMT, GICC, EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (M.-C.V.-M.); (G.P.)
| | - Fabrice Gouilleux
- LNOx, GICC, CNRS ERL 7001, University of Tours, 37000 Tours, France; (M.B.-B.); (M.D.); (N.V.); (O.H.); (F.M.)
- Correspondence: ; Tel.: +33-(2)-47-36-62-91
| |
Collapse
|
43
|
Orlova A, Wagner C, de Araujo ED, Bajusz D, Neubauer HA, Herling M, Gunning PT, Keserű GM, Moriggl R. Direct Targeting Options for STAT3 and STAT5 in Cancer. Cancers (Basel) 2019; 11:E1930. [PMID: 31817042 PMCID: PMC6966570 DOI: 10.3390/cancers11121930] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/22/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022] Open
Abstract
Signal transducer and activator of transcription (STAT)3 and STAT5 are important transcription factors that are able to mediate or even drive cancer progression through hyperactivation or gain-of-function mutations. Mutated STAT3 is mainly associated with large granular lymphocytic T-cell leukemia, whereas mutated STAT5B is associated with T-cell prolymphocytic leukemia, T-cell acute lymphoblastic leukemia and γδ T-cell-derived lymphomas. Hyperactive STAT3 and STAT5 are also implicated in various hematopoietic and solid malignancies, such as chronic and acute myeloid leukemia, melanoma or prostate cancer. Classical understanding of STAT functions is linked to their phosphorylated parallel dimer conformation, in which they induce gene transcription. However, the functions of STAT proteins are not limited to their phosphorylated dimerization form. In this review, we discuss the functions and the roles of unphosphorylated STAT3/5 in the context of chromatin remodeling, as well as the impact of STAT5 oligomerization on differential gene expression in hematopoietic neoplasms. The central involvement of STAT3/5 in cancer has made these molecules attractive targets for small-molecule drug development, but currently there are no direct STAT3/5 inhibitors of clinical grade available. We summarize the development of inhibitors against the SH2 domains of STAT3/5 and discuss their applicability as cancer therapeutics.
Collapse
Affiliation(s)
- Anna Orlova
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| | - Christina Wagner
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| | - Elvin D. de Araujo
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (E.D.d.A.); (P.T.G.)
- Centre for Medicinal Chemistry, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Dávid Bajusz
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (D.B.); (G.M.K.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| | - Marco Herling
- Department I of Internal Medicine, Center for Integrated Oncology (CIO), Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center for Molecular Medicine Cologne (CMMC), Cologne University, 50937 Cologne, Germany;
| | - Patrick T. Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (E.D.d.A.); (P.T.G.)
- Centre for Medicinal Chemistry, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - György M. Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (D.B.); (G.M.K.)
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| |
Collapse
|
44
|
van der Zwet JCG, Cordo' V, Canté-Barrett K, Meijerink JPP. Multi-omic approaches to improve outcome for T-cell acute lymphoblastic leukemia patients. Adv Biol Regul 2019; 74:100647. [PMID: 31523030 DOI: 10.1016/j.jbior.2019.100647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/20/2019] [Accepted: 08/23/2019] [Indexed: 06/10/2023]
Abstract
In the last decade, tremendous progress in curative treatment has been made for T-ALL patients using high-intensive, risk-adapted multi-agent chemotherapy. Further treatment intensification to improve the cure rate is not feasible as it will increase the number of toxic deaths. Hence, about 20% of pediatric patients relapse and often die due to acquired therapy resistance. Personalized medicine is of utmost importance to further increase cure rates and is achieved by targeting specific initiation, maintenance or resistance mechanisms of the disease. Genomic sequencing has revealed mutations that characterize genetic subtypes of many cancers including T-ALL. However, leukemia may have various activated pathways that are not accompanied by the presence of mutations. Therefore, screening for mutations alone is not sufficient to identify all molecular targets and leukemic dependencies for therapeutic inhibition. We review the extent of the driving type A and the secondary type B genomic mutations in pediatric T-ALL that may be targeted by specific inhibitors. Additionally, we review the need for additional screening methods on the transcriptional and protein levels. An integrated 'multi-omic' screening will identify potential targets and biomarkers to establish significant progress in future individualized treatment of T-ALL patients.
Collapse
Affiliation(s)
| | - Valentina Cordo'
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | | |
Collapse
|
45
|
JAK/STAT-Activating Genomic Alterations Are a Hallmark of T-PLL. Cancers (Basel) 2019; 11:cancers11121833. [PMID: 31766351 PMCID: PMC6966610 DOI: 10.3390/cancers11121833] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
T-cell prolymphocytic leukemia (T-PLL) is a rare and poor-prognostic mature T-cell leukemia. Recent studies detected genomic aberrations affecting JAK and STAT genes in T-PLL. Due to the limited number of primary patient samples available, genomic analyses of the JAK/STAT pathway have been performed in rather small cohorts. Therefore, we conducted—via a primary-data based pipeline—a meta-analysis that re-evaluated the genomic landscape of T-PLL. It included all available data sets with sequence information on JAK or STAT gene loci in 275 T-PLL. We eliminated overlapping cases and determined a cumulative rate of 62.1% of cases with mutated JAK or STAT genes. Most frequently, JAK1 (6.3%), JAK3 (36.4%), and STAT5B (18.8%) carried somatic single-nucleotide variants (SNVs), with missense mutations in the SH2 or pseudokinase domains as most prevalent. Importantly, these lesions were predominantly subclonal. We did not detect any strong association between mutations of a JAK or STAT gene with clinical characteristics. Irrespective of the presence of gain-of-function (GOF) SNVs, basal phosphorylation of STAT5B was elevated in all analyzed T-PLL. Fittingly, a significant proportion of genes encoding for potential negative regulators of STAT5B showed genomic losses (in 71.4% of T-PLL in total, in 68.4% of T-PLL without any JAK or STAT mutations). They included DUSP4, CD45, TCPTP, SHP1, SOCS1, SOCS3, and HDAC9. Overall, considering such losses of negative regulators and the GOF mutations in JAK and STAT genes, a total of 89.8% of T-PLL revealed a genomic aberration potentially explaining enhanced STAT5B activity. In essence, we present a comprehensive meta-analysis on the highly prevalent genomic lesions that affect genes encoding JAK/STAT signaling components. This provides an overview of possible modes of activation of this pathway in a large cohort of T-PLL. In light of new advances in JAK/STAT inhibitor development, we also outline translational contexts for harnessing active JAK/STAT signaling, which has emerged as a ‘secondary’ hallmark of T-PLL.
Collapse
|
46
|
Targeting STAT3 and STAT5 in Tumor-Associated Immune Cells to Improve Immunotherapy. Cancers (Basel) 2019; 11:cancers11121832. [PMID: 31766350 PMCID: PMC6966642 DOI: 10.3390/cancers11121832] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/16/2019] [Accepted: 11/18/2019] [Indexed: 02/06/2023] Open
Abstract
Oncogene-induced STAT3-activation is central to tumor progression by promoting cancer cell expression of pro-angiogenic and immunosuppressive factors. STAT3 is also activated in infiltrating immune cells including tumor-associated macrophages (TAM) amplifying immune suppression. Consequently, STAT3 is considered as a target for cancer therapy. However, its interplay with other STAT-family members or transcription factors such as NF-κB has to be considered in light of their concerted regulation of immune-related genes. Here, we discuss new attempts at re-educating immune suppressive tumor-associated macrophages towards a CD8 T cell supporting profile, with an emphasis on the role of STAT transcription factors on TAM functional programs. Recent clinical trials using JAK/STAT inhibitors highlighted the negative effects of these molecules on the maintenance and function of effector/memory T cells. Concerted regulation of STAT3 and STAT5 activation in CD8 T effector and memory cells has been shown to impact their tumor-specific responses including intra-tumor accumulation, long-term survival, cytotoxic activity and resistance toward tumor-derived immune suppression. Interestingly, as an escape mechanism, melanoma cells were reported to impede STAT5 nuclear translocation in both CD8 T cells and NK cells. Ours and others results will be discussed in the perspective of new developments in engineered T cell-based adoptive therapies to treat cancer patients.
Collapse
|
47
|
de Araujo ED, Orlova A, Neubauer HA, Bajusz D, Seo HS, Dhe-Paganon S, Keserű GM, Moriggl R, Gunning PT. Structural Implications of STAT3 and STAT5 SH2 Domain Mutations. Cancers (Basel) 2019; 11:E1757. [PMID: 31717342 PMCID: PMC6895964 DOI: 10.3390/cancers11111757] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 01/13/2023] Open
Abstract
Src Homology 2 (SH2) domains arose within metazoan signaling pathways and are involved in protein regulation of multiple pleiotropic cascades. In signal transducer and activator of transcription (STAT) proteins, SH2 domain interactions are critical for molecular activation and nuclear accumulation of phosphorylated STAT dimers to drive transcription. Sequencing analysis of patient samples has revealed the SH2 domain as a hotspot in the mutational landscape of STAT proteins although the functional impact for the vast majority of these mutations remains poorly characterized. Despite several well resolved structures for SH2 domain-containing proteins, structural data regarding the distinctive STAT-type SH2 domain is limited. Here, we review the unique features of STAT-type SH2 domains in the context of all currently reported STAT3 and STAT5 SH2 domain clinical mutations. The genetic volatility of specific regions in the SH2 domain can result in either activating or deactivating mutations at the same site in the domain, underscoring the delicate evolutionary balance of wild type STAT structural motifs in maintaining precise levels of cellular activity. Understanding the molecular and biophysical impact of these disease-associated mutations can uncover convergent mechanisms of action for mutations localized within the STAT SH2 domain to facilitate the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Elvin D. de Araujo
- Centre for Medicinal Chemistry, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada;
- Department of Chemical & Physical Sciences, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Anna Orlova
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria; (A.O.); (H.A.N.); (R.M.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria; (A.O.); (H.A.N.); (R.M.)
| | - Dávid Bajusz
- Medicinal Chemistry Research Group, Research Center for Natural Sciences, 1117 Budapest, Hungary; (D.B.); (G.M.K.)
| | - Hyuk-Soo Seo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; (H.-S.S.); (S.D.-P.)
- Department of Biological Chemistry, Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Sirano Dhe-Paganon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; (H.-S.S.); (S.D.-P.)
- Department of Biological Chemistry, Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - György M. Keserű
- Medicinal Chemistry Research Group, Research Center for Natural Sciences, 1117 Budapest, Hungary; (D.B.); (G.M.K.)
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, A-1210 Vienna, Austria; (A.O.); (H.A.N.); (R.M.)
| | - Patrick T. Gunning
- Centre for Medicinal Chemistry, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada;
- Department of Chemical & Physical Sciences, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada
| |
Collapse
|
48
|
Maurer B, Kollmann S, Pickem J, Hoelbl-Kovacic A, Sexl V. STAT5A and STAT5B-Twins with Different Personalities in Hematopoiesis and Leukemia. Cancers (Basel) 2019; 11:E1726. [PMID: 31690038 PMCID: PMC6895831 DOI: 10.3390/cancers11111726] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/25/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022] Open
Abstract
The transcription factors STAT5A and STAT5B have essential roles in survival and proliferation of hematopoietic cells-which have been considered largely redundant. Mutations of upstream kinases, copy number gains, or activating mutations in STAT5A, or more frequently in STAT5B, cause altered hematopoiesis and cancer. Interfering with their activity by pharmacological intervention is an up-and-coming therapeutic avenue. Precision medicine requests detailed knowledge of STAT5A's and STAT5B's individual functions. Recent evidence highlights the privileged role for STAT5B over STAT5A in normal and malignant hematopoiesis. Here, we provide an overview on their individual functions within the hematopoietic system.
Collapse
Affiliation(s)
- Barbara Maurer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Sebastian Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Judith Pickem
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Andrea Hoelbl-Kovacic
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| |
Collapse
|
49
|
JAK/STAT Pathway Mutations in T-ALL, Including the STAT5B N642H Mutation, are Sensitive to JAK1/JAK3 Inhibitors. Hemasphere 2019; 3:e313. [PMID: 31976485 PMCID: PMC6924561 DOI: 10.1097/hs9.0000000000000313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/14/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022] Open
|
50
|
Orlova A, Neubauer HA, Moriggl R. The stromal microenvironment provides an escape route from FLT3 inhibitors through the GAS6-AXL-STAT5 axis. Haematologica 2019; 104:1907-1909. [PMID: 31575669 PMCID: PMC6886418 DOI: 10.3324/haematol.2019.225862] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Anna Orlova
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine
- Medical University Vienna, Vienna, Austria
| |
Collapse
|