1
|
Pasquier F, Pegliasco J, Martin JE, Marti S, Plo I. New approaches to standard of care in early-phase myeloproliferative neoplasms: can interferon-α alter the natural history of the disease? Haematologica 2025; 110:850-862. [PMID: 39445431 PMCID: PMC11959252 DOI: 10.3324/haematol.2023.283958] [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: 01/28/2024] [Accepted: 04/15/2024] [Indexed: 10/25/2024] Open
Abstract
The classical BCR::ABL-negative myeloproliferative neoplasms (MPN) include polycythemia vera, essential thrombocythemia, and primary myelofibrosis. They are acquired clonal disorders of hematopoietic stem cells leading to hyperplasia of one or several myeloid lineages. MPN are caused by three main recurrent mutations, JAK2V617F and mutations in the calreticulin (CALR) and thrombopoietin receptor (MPL) genes. Here, we review the general diagnosis, the complications, and the management of MPN. Second, we explain the physiopathology of the natural disease development and its regulation, which contributes to MPN heterogeneity. Thirdly, we describe the new paradigm of MPN development highlighting the early origin of driver mutations, decades before the onset of symptoms, and the consequence of early detection of MPN cases in the general population for prompt diagnosis and better medical management. Finally, we present interferon-α therapy as a potential, early disease-modifying drug after reporting its good hematologic and molecular efficacies in polycythemia vera, essential thrombocythemia, and early myelofibrosis in clinical trials as well as its mechanism of action in pre-clinical studies. As a result, we may expect that, in the future, MPN patients will be diagnosed very early during the course of disease and that new selective therapies under development, such as interferon-α, JAK2V617F inhibitors and CALRmut monoclonal antibodies, will be able to intercept the mutated clones.
Collapse
Affiliation(s)
| | - Jean Pegliasco
- INSERM U1287, Gustave Roussy, Villejuif
- Gustave Roussy, Villejuif
- Université Paris-Cité, Paris, France
| | - Jean-Edouard Martin
- INSERM U1287, Gustave Roussy, Villejuif
- Gustave Roussy, Villejuif
- Université Paris-Cité, Paris, France
| | - Séverine Marti
- INSERM U1287, Gustave Roussy, Villejuif
- Gustave Roussy, Villejuif
- Université Paris-Cité, Paris, France
| | | |
Collapse
|
2
|
Thiele J, Kvasnicka HM, Gianelli U, Arber DA, Tefferi A, Vannucchi AM, Barbui T, Orazi A. Evolution of WHO diagnostic criteria in "Classical Myeloproliferative Neoplasms" compared with the International Consensus Classification. Blood Cancer J 2025; 15:31. [PMID: 40038244 PMCID: PMC11880409 DOI: 10.1038/s41408-025-01235-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/15/2025] [Accepted: 02/12/2025] [Indexed: 03/06/2025] Open
Abstract
A lively discussion persists regarding the diagnostic criteria for essential thrombocythemia (ET), primary myelofibrosis (PMF) and polycythemia vera (PV), particularly in relation to early/pre-fibrotic myelofibrosis (pre-PMF), a disease entity initially introduced in 2001 by the 3rd edition of the World Health Organization (WHO) classification. The definition and criteria used to diagnose pre-PMF have been progressively modified over time. The most update definition of pre-PMF can be found in the International Consensus Classification (ICC) published in 2022. An updated largely similar definition is also incorporated in the recently published 5th edition of WHO classification (2024). Diagnostic criteria for ET have undergone changes up to 2016/17 for the revised 4th edition of the WHO. In particular the threshold value for platelets were lowered and the important discrimination between "true" and "false" ET (in reality pre-PMF) been widely acknowledged. To avoid misdiagnose in early phase PV, the criteria for gender-adjusted thresholds for hemoglobin/ hematocrit have been lowered and the identification of an appropriate bone marrow (BM) morphology was upgraded as a major diagnostic criterion. Given the prominent role of morphology in MPN-related diagnostic algorithms, the diagnostic adequacy of the BM biopsy (sample procurement and proper laboratory handling) as emphasized in former WHO editions and in the ICC, was not addressed by the WHO 5th. The essential role of genetic markers is recognized by both classifications. A comparison between the revised 4th edition WHO classification and the ICC versus the WHO 5th reveals no significant differences, with the exception of the occurrence of leukoerythroblastosis in pre-PMF considered by the latter as one of the minor diagnostic criteria which seems unwarranted. In contrast to the revised 4th edition, the majority of the microscopic images used for the WHO 5th due to their low magnification and poor technique, do not highlight the diagnosis differences among these entities.
Collapse
Affiliation(s)
- Jürgen Thiele
- Institute of Pathology, University of Cologne, Cologne, Germany
| | | | - Umberto Gianelli
- University of Milan, Department of Health Sciences and S.C. Anatomia Patologica, ASST Santi Paolo e Carlo, Milan, Italy
| | - Daniel A Arber
- Department of Pathology, University of Chicago, Chicago, IL, US
| | | | - Alessandro M Vannucchi
- CRIMM-Centro Ricerca e Innovazione delle Malattie Mieloproliferative, Azienda Ospedaliera-Universitaria Careggi, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Tiziano Barbui
- FROM Research Foundation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Attilio Orazi
- Department of Pathology, Texas Tech University Health Sciences Center, El Paso, TX, US.
| |
Collapse
|
3
|
Kaur A, Venkatesan A, Kandarpa M, Talpaz M, Raghavan M. Lysosomal degradation targets mutant calreticulin and the thrombopoietin receptor in myeloproliferative neoplasms. Blood Adv 2024; 8:3372-3387. [PMID: 38640435 PMCID: PMC11255115 DOI: 10.1182/bloodadvances.2023011432] [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: 08/14/2023] [Revised: 03/24/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024] Open
Abstract
ABSTRACT Somatic mutants of calreticulin (CRT) drive myeloproliferative neoplasms (MPNs) via binding to the thrombopoietin receptor (MPL) and aberrant activation of the JAK/STAT pathway. Compared with healthy donors, platelets from mutant CRT-expressing patients with MPN display low cell surface MPL. Additionally, coexpression of MPL with an MPN-linked CRT mutant (CRTDel52) reduces cell surface MPL, suggesting that CRTDel52 may induce MPL degradation. We show that lysosomal degradation is relevant to the turnover of CRTDel52 and MPL. Furthermore, CRTDel52 increases the lysosomal localization and degradation of MPL. Mammalian target of rapamycin (mTOR) inhibitors reduce cellular CRTDel52 and MPL, secreted CRTDel52 levels, and impair CRTDel52-mediated cell proliferation. mTOR inhibition also reduces colony formation and differentiation of CD34+ cells from patients with MPN but not from healthy donors. Together, these findings indicate that low-surface MPL is a biomarker of mutant CRT-mediated MPN and that induced degradation of CRTDel52 and MPL is an avenue for therapeutic intervention.
Collapse
Affiliation(s)
- Amanpreet Kaur
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Arunkumar Venkatesan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Malathi Kandarpa
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Moshe Talpaz
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Malini Raghavan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
4
|
Vermeersch G, Proost P, Struyf S, Gouwy M, Devos T. CXCL8 and its cognate receptors CXCR1/CXCR2 in primary myelofibrosis. Haematologica 2024; 109:2060-2072. [PMID: 38426279 PMCID: PMC11215396 DOI: 10.3324/haematol.2023.284921] [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: 12/27/2023] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
BCR::ABL1 negative myeloproliferative neoplasms (MPN) form a distinct group of hematologic malignancies characterized by sustained proliferation of cells from multiple myeloid lineages. With a median survival of 16-35 months in patients with high-risk disease, primary myelofibrosis (PMF) is considered the most aggressive entity amongst all BCR::ABL1 MPN. Additionally, for a significant subset of patients, MPN evolve into secondary acute myeloid leukemia (AML), which has an even poorer prognosis compared to de novo AML. As the exact mechanisms of disease development and progression remain to be elucidated, current therapeutic approaches fail to prevent disease progression or transformation into secondary AML. As each MPN entity is characterized by sustained activation of various immune cells and raised cytokine concentrations within bone marrow (BM) and peripheral blood (PB), MPN may be considered to be typical inflammation-related malignancies. However, the exact role and consequences of increased cytokine concentrations within BM and PB plasma has still not been completely established. Up-regulated cytokines can stimulate cellular proliferation, or contribute to the development of an inflammation-related BM niche resulting in genotoxicity and thereby supporting mutagenesis. The neutrophil chemoattractant CXCL8 is of specific interest as its concentration is increased within PB and BM plasma of patients with PMF. Increased concentration of CXCL8 negatively correlates with overall survival. Furthermore, blockage of the CXCR1/2 axis appears to be able to reduce BM fibrosis and megakaryocyte dysmorphia in murine models. In this review, we summarize available evidence on the role of the CXCL8-CXCR1/2 axis within the pathogenesis of PMF, and discuss potential therapeutic modalities targeting either CXCL8 or its cognate receptors CXCR1/2.
Collapse
Affiliation(s)
- Gael Vermeersch
- Department of Hematology, University Hospitals Leuven, 3000, Leuven, Belgium; Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000, Leuven
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000, Leuven
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000, Leuven
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000, Leuven
| | - Timothy Devos
- Department of Hematology, University Hospitals Leuven, 3000, Leuven, Belgium; Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, 3000, Leuven.
| |
Collapse
|
5
|
Behrens K, Kauppi M, Viney EM, Kueh AJ, Hyland CD, Willson TA, Salleh L, de Graaf CA, Babon JJ, Herold MJ, Nicola NA, Alexander WS. Differential in vivo roles of Mpl cytoplasmic tyrosine residues in murine hematopoiesis and myeloproliferative disease. Leukemia 2024; 38:1342-1352. [PMID: 38491305 PMCID: PMC11147766 DOI: 10.1038/s41375-024-02219-5] [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: 07/18/2023] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Thrombopoietin (Tpo), which binds to its specific receptor, the Mpl protein, is the major cytokine regulator of megakaryopoiesis and circulating platelet number. Tpo binding to Mpl triggers activation of Janus kinase 2 (Jak2) and phosphorylation of the receptor, as well as activation of several intracellular signalling cascades that mediate cellular responses. Three tyrosine (Y) residues in the C-terminal region of the Mpl intracellular domain have been implicated as sites of phosphorylation required for regulation of major Tpo-stimulated signalling pathways: Mpl-Y565, Mpl-Y599 and Mpl-Y604. Here, we have introduced mutations in the mouse germline and report a consistent physiological requirement for Mpl-Y599, mutation of which resulted in thrombocytopenia, deficient megakaryopoiesis, low hematopoietic stem cell (HSC) number and function, and attenuated responses to myelosuppression. We further show that in models of myeloproliferative neoplasms (MPN), where Mpl is required for pathogenesis, thrombocytosis was dependent on intact Mpl-Y599. In contrast, Mpl-Y565 was required for negative regulation of Tpo responses; mutation of this residue resulted in excess megakaryopoiesis at steady-state and in response to myelosuppression, and exacerbated thrombocytosis associated with MPN.
Collapse
Affiliation(s)
- Kira Behrens
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Maria Kauppi
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Elizabeth M Viney
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Andrew J Kueh
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, 3084, Australia
| | - Craig D Hyland
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Tracy A Willson
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Liam Salleh
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Carolyn A de Graaf
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jeffrey J Babon
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Marco J Herold
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, 3084, Australia
| | - Nicos A Nicola
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Warren S Alexander
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
6
|
Tefferi A, Vannucchi AM, Barbui T. Essential thrombocythemia: 2024 update on diagnosis, risk stratification, and management. Am J Hematol 2024; 99:697-718. [PMID: 38269572 DOI: 10.1002/ajh.27216] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/26/2024]
Abstract
OVERVIEW Essential thrombocythemia is a Janus kinase 2 (JAK2) mutation-prevalent myeloproliferative neoplasm characterized by clonal thrombocytosis; clinical course is often indolent but might be interrupted by thrombotic or hemorrhagic complications, microcirculatory symptoms (e.g., headaches, lightheadedness, and acral paresthesias), and, less frequently, by disease transformation into myelofibrosis (MF) or acute myeloid leukemia. DIAGNOSIS In addition to thrombocytosis (platelets ≥450 × 109 /L), formal diagnosis requires the exclusion of other myeloid neoplasms, including prefibrotic MF, polycythemia vera, chronic myeloid leukemia, and myelodysplastic syndromes with ring sideroblasts and thrombocytosis. Bone marrow morphology typically shows increased number of mature-appearing megakaryocytes distributed in loose clusters. GENETICS Approximately 80% of patients express myeloproliferative neoplasm driver mutations (JAK2, CALR, MPL), in a mutually exclusive manner; in addition, about 50% harbor other mutations, the most frequent being TET2 (9%-11%), ASXL1 (7%-20%), DNMT3A (7%), and SF3B1 (5%). Abnormal karyotype is seen in <10% of patients and includes +9/20q-/13q-. SURVIVAL AND PROGNOSIS Life expectancy is less than that of the control population. Median survival is approximately 18 years but exceeds >35 years in younger patients. The triple A survival risk model, based on Age, Absolute neutrophil count, and Absolute lymphocyte count, effectively delineates high-, intermediate-1-, intermediate-2-, and low-risk disease with corresponding median survivals of 8, 14, 21, and 47 years. RISK FACTORS FOR THROMBOSIS Four risk categories are considered: very low (age ≤60 years, no thrombosis history, JAK2 wild-type), low (same as very low but JAK2 mutation present), intermediate (same as low but age >60 years), and high (thrombosis history or age >60 years with JAK2 mutation). MUTATIONS AND PROGNOSIS MPL and CALR-1 mutations have been associated with increased risk of MF transformation; spliceosome with inferior overall and MF-free survival; TP53 with leukemic transformation, and JAK2V617F with thrombosis. Leukemic transformation rate at 10 years is <1% but might be higher in JAK2-mutated patients with extreme thrombocytosis and those with abnormal karyotype. TREATMENT The main goal of therapy is to prevent thrombosis. In this regard, once-daily low-dose aspirin is advised for all patients and twice daily for low-risk disease. Cytoreductive therapy is advised for high-risk and optional for intermediate-risk disease. First-line cytoreductive drugs of choice are hydroxyurea and pegylated interferon-α and second-line busulfan. ADDITIONAL CONTENT The current review includes specific treatment strategies in the context of extreme thrombocytosis, pregnancy, splanchnic vein thrombosis, perioperative care, and post-essential thrombocythemia MF, as well as new investigational drugs.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Alessandro Maria Vannucchi
- CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence, AOU Careggi, Florence, Italy
| | - Tiziano Barbui
- Research Foundation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| |
Collapse
|
7
|
Barosi G, Campanelli R, Catarsi P, Abbà C, Carolei A, Massa M, Gale RP, Rosti V. Type 1 CALR mutation allele frequency correlates with CD34/CXCR4 expression in myelofibrosis-type megakaryocyte dysplasia: A mechanism of disease progression? Blood Cancer J 2024; 14:18. [PMID: 38253566 PMCID: PMC10803778 DOI: 10.1038/s41408-024-00991-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Affiliation(s)
- Giovanni Barosi
- Center for the Study of Myelofibrosis, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo Foundation, Pavia, Italy.
| | - Rita Campanelli
- Center for the Study of Myelofibrosis, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo Foundation, Pavia, Italy
| | - Paolo Catarsi
- Center for the Study of Myelofibrosis, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo Foundation, Pavia, Italy
| | - Carlotta Abbà
- General Medicine 2, Center for Sistemic Amyloidosis and High Complexity Diseases, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo Foundation, Pavia, Italy
| | - Adriana Carolei
- Center for the Study of Myelofibrosis, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo Foundation, Pavia, Italy
| | - Margherita Massa
- General Medicine 2, Center for Sistemic Amyloidosis and High Complexity Diseases, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo Foundation, Pavia, Italy
| | - Robert Peter Gale
- Centre for Haematology Research, Imperial College London, London, UK
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico S. Matteo Foundation, Pavia, Italy
| |
Collapse
|
8
|
Papadopoulos N, Pristavec A, Nédélec A, Levy G, Staerk J, Constantinescu SN. Modulation of human thrombopoietin receptor conformations uncouples JAK2 V617F-driven activation from cytokine-induced stimulation. Blood 2023; 142:1818-1830. [PMID: 37616564 DOI: 10.1182/blood.2022019580] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023] Open
Abstract
The thrombopoietin receptor (TpoR) plays a central role in myeloproliferative neoplasms (MPNs). Mutations in JAK2, calreticulin, or TpoR itself drive the constitutive activation of TpoR and uncontrolled proliferation and differentiation of hematopoietic stem cells and progenitors. The JAK2 V617F mutation is responsible for most MPNs, and all driver mutants induce pathologic TpoR activation. Existing therapeutic strategies have focused on JAK2 kinase inhibitors that are unable to differentiate between the mutated MPN clone and healthy cells. Surprisingly, the targeting of TpoR itself has remained poorly explored despite its central role in pathology. Here, we performed a comprehensive characterization of human TpoR activation under physiological and pathological conditions, focusing on the JAK2 V617F mutant. Using a system of controlled dimerization of the transmembrane and cytosolic domains of TpoR, we discovered that human TpoR (hTpoR) adopts different dimeric conformations upon Tpo-induced vs JAK2 V617F-mediated activation. We identified the amino acids and specific dimeric conformation of hTpoR responsible for activation in complex with JAK2 V617F and confirmed our findings in the full-length receptor context in hematopoietic cell lines and primary bone marrow cells. Remarkably, we found that the modulation of hTpoR conformations by point mutations allowed for specific inhibition of JAK2 V617F-driven activation without affecting Tpo-induced signaling. Our results demonstrate that modulation of the hTpoR conformation is a viable therapeutic strategy for JAK2 V617F-positive MPNs and set the path for novel drug development by identifying precise residues of hTpoR involved in JAK2 V617F-specific activation.
Collapse
Affiliation(s)
- Nicolas Papadopoulos
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology Department, Walloon Excellence Research Institute, Wavre, Belgium
| | - Ajda Pristavec
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Audrey Nédélec
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology Department, Walloon Excellence Research Institute, Wavre, Belgium
| | - Gabriel Levy
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology Department, Walloon Excellence Research Institute, Wavre, Belgium
| | - Judith Staerk
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, Brussels, Belgium
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, University of Oslo, Oslo, Norway
| | - Stefan N Constantinescu
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology Department, Walloon Excellence Research Institute, Wavre, Belgium
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research, Oxford University, Oxford, United Kingdom
| |
Collapse
|
9
|
Willekens C, Laplane L, Dagher T, Benlabiod C, Papadopoulos N, Lacout C, Rameau P, Catelain C, Alfaro A, Edmond V, Signolle N, Marchand V, Droin N, Hoogenboezem R, Schneider RK, Penson A, Abdel-Wahab O, Giraudier S, Pasquier F, Marty C, Plo I, Villeval JL, Constantinescu SN, Porteu F, Vainchenker W, Solary E. SRSF2-P95H decreases JAK/STAT signaling in hematopoietic cells and delays myelofibrosis development in mice. Leukemia 2023:10.1038/s41375-023-01878-0. [PMID: 37100881 DOI: 10.1038/s41375-023-01878-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 04/28/2023]
Abstract
Heterozygous mutation targeting proline 95 in Serine/Arginine-rich Splicing Factor 2 (SRSF2) is associated with V617F mutation in Janus Activated Kinase 2 (JAK2) in some myeloproliferative neoplasms (MPNs), most commonly primary myelofibrosis. To explore the interaction of Srsf2P95H with Jak2V617F, we generated Cre-inducible knock-in mice expressing these mutants under control of the stem cell leukemia (Scl) gene promoter. In transplantation experiments, Srsf2P95H unexpectedly delayed myelofibrosis induced by Jak2V617F and decreased TGFβ1 serum level. Srsf2P95H reduced the competitiveness of transplanted Jak2V617F hematopoietic stem cells while preventing their exhaustion. RNA sequencing of sorted megakaryocytes identified an increased number of splicing events when the two mutations were combined. Focusing on JAK/STAT pathway, Jak2 exon 14 skipping was promoted by Srsf2P95H, an event detected in patients with JAK2V617F and SRSF2P95 co-mutation. The skipping event generates a truncated inactive JAK2 protein. Accordingly, Srsf2P95H delays myelofibrosis induced by the thrombopoietin receptor agonist Romiplostim in Jak2 wild-type animals. These results unveil JAK2 exon 14 skipping promotion as a strategy to reduce JAK/STAT signaling in pathological conditions.
Collapse
Affiliation(s)
- Christophe Willekens
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Département d'hématologie, Gustave Roussy Cancer Campus, Villejuif, France
| | - Lucie Laplane
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Institut d'Histoire et Philosophie des Sciences et des Techniques, Université Paris I Panthéon-Sorbonne, Paris, France
| | - Tracy Dagher
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Camelia Benlabiod
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Institut d'Histoire et Philosophie des Sciences et des Techniques, Université Paris I Panthéon-Sorbonne, Paris, France
| | - Nicolas Papadopoulos
- Ludwig Institute for Cancer Research Brussels, Brussels, Belgium
- Université catholique de Louvain and de Duve Institute, Brussels, Belgium
| | | | | | | | | | - Valérie Edmond
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Valentine Marchand
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Nathalie Droin
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Remco Hoogenboezem
- Department of Hematology, Erasmus University, Rotterdam, The Netherlands
| | - Rebekka K Schneider
- Department of Hematology, Erasmus University, Rotterdam, The Netherlands
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, RWTH Aachen University, Aachen, Germany
| | - Alex Penson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Florence Pasquier
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Département d'hématologie, Gustave Roussy Cancer Campus, Villejuif, France
| | - Caroline Marty
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Isabelle Plo
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Jean-Luc Villeval
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research Brussels, Brussels, Belgium
- Université catholique de Louvain and de Duve Institute, Brussels, Belgium
- WELBIO department, WEL Research Institute, Wavre, Belgium
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | - Françoise Porteu
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - William Vainchenker
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Eric Solary
- INSERM U1287, Gustave Roussy Cancer Campus, Villejuif, France.
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.
- Département d'hématologie, Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
10
|
How J, Garcia JS, Mullally A. Biology and therapeutic targeting of molecular mechanisms in MPNs. Blood 2023; 141:1922-1933. [PMID: 36534936 PMCID: PMC10163317 DOI: 10.1182/blood.2022017416] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell disorders characterized by activated Janus kinase (JAK)-signal transducer and activator of transcription signaling. As a result, JAK inhibitors have been the standard therapy for treatment of patients with myelofibrosis (MF). Although currently approved JAK inhibitors successfully ameliorate MPN-related symptoms, they are not known to substantially alter the MF disease course. Similarly, in essential thrombocythemia and polycythemia vera, treatments are primarily aimed at reducing the risk of cardiovascular and thromboembolic complications, with a watchful waiting approach often used in patients who are considered to be at a lower risk for thrombosis. However, better understanding of MPN biology has led to the development of rationally designed therapies, with the goal of not only addressing disease complications but also potentially modifying disease course. We review the most recent data elucidating mechanisms of disease pathogenesis and highlight emerging therapies that target MPN on several biologic levels, including JAK2-mutant MPN stem cells, JAK and non-JAK signaling pathways, mutant calreticulin, and the inflammatory bone marrow microenvironment.
Collapse
Affiliation(s)
- Joan How
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Jacqueline S. Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute, Cambridge, MA
| |
Collapse
|
11
|
Desikan H, Kaur A, Pogozheva ID, Raghavan M. Effects of calreticulin mutations on cell transformation and immunity. J Cell Mol Med 2023; 27:1032-1044. [PMID: 36916035 PMCID: PMC10098294 DOI: 10.1111/jcmm.17713] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are cancers involving dysregulated production and function of myeloid lineage hematopoietic cells. Among MPNs, Essential thrombocythemia (ET), Polycythemia Vera (PV) and Myelofibrosis (MF), are driven by mutations that activate the JAK-STAT signalling pathway. Somatic mutations of calreticulin (CRT), an endoplasmic reticulum (ER)-localized lectin chaperone, are driver mutations in approximately 25% of ET and 35% of MF patients. The MPN-linked mutant CRT proteins have novel frameshifted carboxy-domain sequences and lack an ER retention motif, resulting in their secretion. Wild type CRT is a regulator of ER calcium homeostasis and plays a key role in the assembly of major histocompatibility complex (MHC) class I molecules, which are the ligands for antigen receptors of CD8+ T cells. Mutant CRT-linked oncogenesis results from the dysregulation of calcium signalling in cells and the formation of stable complexes of mutant CRT with myeloproliferative leukemia (MPL) protein, followed by downstream activation of the JAK-STAT signalling pathway. The intricate participation of CRT in ER protein folding, calcium homeostasis and immunity suggests the involvement of multiple mechanisms of mutant CRT-linked oncogenesis. In this review, we highlight recent findings related to the role of MPN-linked CRT mutations in the dysregulation of calcium homeostasis, MPL activation and immunity.
Collapse
Affiliation(s)
- Harini Desikan
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Amanpreet Kaur
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Irina D. Pogozheva
- Department of Medicinal ChemistryCollege of Pharmacy, University of MichiganAnn ArborMichiganUSA
| | - Malini Raghavan
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
12
|
Pecquet C, Papadopoulos N, Balligand T, Chachoua I, Tisserand A, Vertenoeil G, Nédélec A, Vertommen D, Roy A, Marty C, Nivarthi H, Defour JP, El-Khoury M, Hug E, Majoros A, Xu E, Zagrijtschuk O, Fertig TE, Marta DS, Gisslinger H, Gisslinger B, Schalling M, Casetti I, Rumi E, Pietra D, Cavalloni C, Arcaini L, Cazzola M, Komatsu N, Kihara Y, Sunami Y, Edahiro Y, Araki M, Lesyk R, Buxhofer-Ausch V, Heibl S, Pasquier F, Havelange V, Plo I, Vainchenker W, Kralovics R, Constantinescu SN. Secreted mutant calreticulins as rogue cytokines in myeloproliferative neoplasms. Blood 2023; 141:917-929. [PMID: 36356299 PMCID: PMC10651872 DOI: 10.1182/blood.2022016846] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 10/05/2022] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Mutant calreticulin (CALR) proteins resulting from a -1/+2 frameshifting mutation of the CALR exon 9 carry a novel C-terminal amino acid sequence and drive the development of myeloproliferative neoplasms (MPNs). Mutant CALRs were shown to interact with and activate the thrombopoietin receptor (TpoR/MPL) in the same cell. We report that mutant CALR proteins are secreted and can be found in patient plasma at levels up to 160 ng/mL, with a mean of 25.64 ng/mL. Plasma mutant CALR is found in complex with soluble transferrin receptor 1 (sTFR1) that acts as a carrier protein and increases mutant CALR half-life. Recombinant mutant CALR proteins bound and activated the TpoR in cell lines and primary megakaryocytic progenitors from patients with mutated CALR in which they drive thrombopoietin-independent colony formation. Importantly, the CALR-sTFR1 complex remains functional for TpoR activation. By bioluminescence resonance energy transfer assay, we show that mutant CALR proteins produced in 1 cell can specifically interact in trans with the TpoR on a target cell. In comparison with cells that only carry TpoR, cells that carry both TpoR and mutant CALR are hypersensitive to exogenous mutant CALR proteins and respond to levels of mutant CALR proteins similar to those in patient plasma. This is consistent with CALR-mutated cells that expose TpoR carrying immature N-linked sugars at the cell surface. Thus, secreted mutant CALR proteins will act more specifically on the MPN clone. In conclusion, a chaperone, CALR, can turn into a rogue cytokine through somatic mutation of its encoding gene.
Collapse
Affiliation(s)
- Christian Pecquet
- Ludwig Cancer Research, Brussels, Belgium
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
| | - Nicolas Papadopoulos
- Ludwig Cancer Research, Brussels, Belgium
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
| | - Thomas Balligand
- Ludwig Cancer Research, Brussels, Belgium
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
| | - Ilyas Chachoua
- Ludwig Cancer Research, Brussels, Belgium
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Amandine Tisserand
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Université Paris Cité, UMR 1287, Gustave Roussy, Villejuif, France
- UMR 1287, Gustave Roussy, Villejuif, France
| | - Gaëlle Vertenoeil
- Ludwig Cancer Research, Brussels, Belgium
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
| | - Audrey Nédélec
- Ludwig Cancer Research, Brussels, Belgium
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
| | - Didier Vertommen
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
| | - Anita Roy
- Ludwig Cancer Research, Brussels, Belgium
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
| | - Caroline Marty
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- UMR 1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France
| | - Harini Nivarthi
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Jean-Philippe Defour
- Ludwig Cancer Research, Brussels, Belgium
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
| | - Mira El-Khoury
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- UMR 1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France
| | - Eva Hug
- MyeloPro Diagnostics and Research GmbH, Vienna, Austria
| | | | - Erica Xu
- MyeloPro Diagnostics and Research GmbH, Vienna, Austria
| | | | | | - Daciana S. Marta
- Ultrastructural Pathology Lab and Bioimaging, Institute of Pathology Victor Babeș, Bucharest, Romania
| | - Heinz Gisslinger
- Division of Hematology and Blood Coagulation, Department of Internal Medicine I, Division of Hematology and Blood Coagulation, Medical University of Vienna, Vienna, Austria
| | - Bettina Gisslinger
- Division of Hematology and Blood Coagulation, Department of Internal Medicine I, Division of Hematology and Blood Coagulation, Medical University of Vienna, Vienna, Austria
| | - Martin Schalling
- Division of Hematology and Blood Coagulation, Department of Internal Medicine I, Division of Hematology and Blood Coagulation, Medical University of Vienna, Vienna, Austria
| | - Ilaria Casetti
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Elisa Rumi
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Daniela Pietra
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Chiara Cavalloni
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Luca Arcaini
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Mario Cazzola
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Norio Komatsu
- Department of Hematology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yoshihiko Kihara
- Department of Hematology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yoshitaka Sunami
- Department of Hematology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yoko Edahiro
- Department of Hematology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Marito Araki
- Department of Transfusion Medicine and Stem Cell Regulation, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Roman Lesyk
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Rzeszow, Poland
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Veronika Buxhofer-Ausch
- Department of Internal Medicine I with Hematology, Ordensklinikum Linz Elisabethinen, Stem Cell Transplantation Hemostaseology and Medical Oncology, Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Sonja Heibl
- Department of Internal Medicine IV, Klinikum Wels-Grieskirchen, Wels, Austria
| | - Florence Pasquier
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Université Paris Cité, UMR 1287, Gustave Roussy, Villejuif, France
- UMR 1287, Gustave Roussy, Villejuif, France
- Department of Hematology, Gustave Roussy, Villejuif, France
| | - Violaine Havelange
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
- Department of Hematology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Isabelle Plo
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- UMR 1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France
| | - William Vainchenker
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- UMR 1287, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France
| | - Robert Kralovics
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Stefan N. Constantinescu
- Ludwig Cancer Research, Brussels, Belgium
- Université Catholique de Louvain and de Duve Institute, SIGN Unit, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| |
Collapse
|
13
|
Foßelteder J, Pabst G, Sconocchia T, Schlacher A, Auinger L, Kashofer K, Beham-Schmid C, Trajanoski S, Waskow C, Schöll W, Sill H, Zebisch A, Wölfler A, Thomas D, Reinisch A. Human gene-engineered calreticulin mutant stem cells recapitulate MPN hallmarks and identify targetable vulnerabilities. Leukemia 2023; 37:843-853. [PMID: 36813992 PMCID: PMC10079532 DOI: 10.1038/s41375-023-01848-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023]
Abstract
Calreticulin (CALR) mutations present the main oncogenic drivers in JAK2 wildtype (WT) myeloproliferative neoplasms (MPN), including essential thrombocythemia and myelofibrosis, where mutant (MUT) CALR is increasingly recognized as a suitable mutation-specific drug target. However, our current understanding of its mechanism-of-action is derived from mouse models or immortalized cell lines, where cross-species differences, ectopic over-expression and lack of disease penetrance are hampering translational research. Here, we describe the first human gene-engineered model of CALR MUT MPN using a CRISPR/Cas9 and adeno-associated viral vector-mediated knock-in strategy in primary human hematopoietic stem and progenitor cells (HSPCs) to establish a reproducible and trackable phenotype in vitro and in xenografted mice. Our humanized model recapitulates many disease hallmarks: thrombopoietin-independent megakaryopoiesis, myeloid-lineage skewing, splenomegaly, bone marrow fibrosis, and expansion of megakaryocyte-primed CD41+ progenitors. Strikingly, introduction of CALR mutations enforced early reprogramming of human HSPCs and the induction of an endoplasmic reticulum stress response. The observed compensatory upregulation of chaperones revealed novel mutation-specific vulnerabilities with preferential sensitivity of CALR mutant cells to inhibition of the BiP chaperone and the proteasome. Overall, our humanized model improves purely murine models and provides a readily usable basis for testing of novel therapeutic strategies in a human setting.
Collapse
Affiliation(s)
- Johannes Foßelteder
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Gabriel Pabst
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria.,Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria.,Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Tommaso Sconocchia
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Angelika Schlacher
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Lisa Auinger
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Karl Kashofer
- Diagnostic & Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Medical University of Graz, Graz, Austria
| | - Claudia Waskow
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany.,Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich-Schiller-University, Jena, Germany
| | - Wolfgang Schöll
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Heinz Sill
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Armin Zebisch
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria.,Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Albert Wölfler
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Daniel Thomas
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Andreas Reinisch
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria.
| |
Collapse
|
14
|
Inferring the initiation and development of myeloproliferative neoplasms. Proc Natl Acad Sci U S A 2022; 119:e2120374119. [PMID: 36083966 PMCID: PMC9478641 DOI: 10.1073/pnas.2120374119] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The developmental history of blood cancer begins with mutation acquisition and the resulting malignant clone expansion. The two most prevalent driver mutations found in myeloproliferative neoplasms-JAK2V617F and CALRm-occur in hematopoietic stem cells, which are highly complex to observe in vivo. To circumvent this difficulty, we propose a method relying on mathematical modeling and statistical inference to determine disease initiation and dynamics. Our findings suggest that CALRm mutations tend to occur later in life than JAK2V617F. Our results confirm the higher proliferative advantage of the CALRm malignant clone compared to JAK2V617F. Furthermore, we illustrate how mathematical modeling and Bayesian inference can be used for setting up early screening strategies.
Collapse
|
15
|
Tefferi A, Gangat N, Pardanani A, Crispino JD. Myelofibrosis: Genetic Characteristics and the Emerging Therapeutic Landscape. Cancer Res 2022; 82:749-763. [PMID: 34911786 PMCID: PMC9306313 DOI: 10.1158/0008-5472.can-21-2930] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/18/2021] [Accepted: 11/30/2021] [Indexed: 01/07/2023]
Abstract
Primary myelofibrosis (PMF) is one of three myeloproliferative neoplasms (MPN) that are morphologically and molecularly inter-related, the other two being polycythemia vera (PV) and essential thrombocythemia (ET). MPNs are characterized by JAK-STAT-activating JAK2, CALR, or MPL mutations that give rise to stem cell-derived clonal myeloproliferation, which is prone to leukemic and, in case of PV and ET, fibrotic transformation. Abnormal megakaryocyte proliferation is accompanied by bone marrow fibrosis and characterizes PMF, while the clinical phenotype is pathogenetically linked to ineffective hematopoiesis and aberrant cytokine expression. Among MPN-associated driver mutations, type 1-like CALR mutation has been associated with favorable prognosis in PMF, while ASXL1, SRSF2, U2AF1-Q157, EZH2, CBL, and K/NRAS mutations have been shown to be prognostically detrimental. Such information has enabled development of exclusively genetic (GIPSS) and clinically integrated (MIPSSv2) prognostic models that facilitate individualized treatment decisions. Allogeneic stem cell transplantation remains the only treatment modality in MF with the potential to prolong survival, whereas drug therapy, including JAK2 inhibitors, is directed mostly at the inflammatory component of the disease and is therefore palliative in nature. Similarly, disease-modifying activity remains elusive for currently available investigational drugs, while their additional value in symptom management awaits controlled confirmation. There is a need for genetic characterization of clinical observations followed by in vitro and in vivo preclinical studies that will hopefully identify therapies that target the malignant clone in MF to improve patient outcomes.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.,Corresponding Author: Ayalew Tefferi, Division of Hematology, Department of Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905. Phone: 507-284-2511; Fax: 507-266-4972; E-mail:
| | - Naseema Gangat
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Animesh Pardanani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - John D. Crispino
- Division of Experimental Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
16
|
Mosca M, Hermange G, Tisserand A, Noble R, Marzac C, Marty C, Le Sueur C, Campario H, Vertenoeil G, El-Khoury M, Catelain C, Rameau P, Gella C, Lenglet J, Casadevall N, Favier R, Solary E, Cassinat B, Kiladjian JJ, Constantinescu SN, Pasquier F, Hochberg ME, Raslova H, Villeval JL, Girodon F, Vainchenker W, Cournède PH, Plo I. Inferring the dynamics of mutated hematopoietic stem and progenitor cells induced by IFNα in myeloproliferative neoplasms. Blood 2021; 138:2231-2243. [PMID: 34407546 PMCID: PMC8641097 DOI: 10.1182/blood.2021010986] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 07/15/2021] [Indexed: 11/20/2022] Open
Abstract
Classical BCR-ABL-negative myeloproliferative neoplasms (MPNs) are clonal disorders of hematopoietic stem cells (HSCs) caused mainly by recurrent mutations in genes encoding JAK2 (JAK2), calreticulin (CALR), or the thrombopoietin receptor (MPL). Interferon α (IFNα) has demonstrated some efficacy in inducing molecular remission in MPNs. To determine factors that influence molecular response rate, we evaluated the long-term molecular efficacy of IFNα in patients with MPN by monitoring the fate of cells carrying driver mutations in a prospective observational and longitudinal study of 48 patients over more than 5 years. We measured the clonal architecture of early and late hematopoietic progenitors (84 845 measurements) and the global variant allele frequency in mature cells (409 measurements) several times per year. Using mathematical modeling and hierarchical Bayesian inference, we further inferred the dynamics of IFNα-targeted mutated HSCs. Our data support the hypothesis that IFNα targets JAK2V617F HSCs by inducing their exit from quiescence and differentiation into progenitors. Our observations indicate that treatment efficacy is higher in homozygous than heterozygous JAK2V617F HSCs and increases with high IFNα dose in heterozygous JAK2V617F HSCs. We also found that the molecular responses of CALRm HSCs to IFNα were heterogeneous, varying between type 1 and type 2 CALRm, and a high dose of IFNα correlates with worse outcomes. Our work indicates that the long-term molecular efficacy of IFNα implies an HSC exhaustion mechanism and depends on both the driver mutation type and IFNα dose.
Collapse
Affiliation(s)
- Matthieu Mosca
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| | - Gurvan Hermange
- Université Paris-Saclay, CentraleSupélec, Laboratory MICS (Laboratory of Applied Mathematics and Computer Science), Gif-sur-Yvette, France
| | - Amandine Tisserand
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Laboratoire d'Excellence GR-Ex, Paris, France
- Université de Paris, Paris, France
| | - Robert Noble
- Department of Biosciences and Engineering, ETH Zurich, Basel, Switzerland
- Institut des Sciences de l'Evolution, University of Montpellier, Montpellier, France
- Institute of Evolutionary Biology and Environmental Studies (IEU), University of Zurich, Zurich, Switzerland
- University of London, London, United Kingdom
| | - Christophe Marzac
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire d'Immuno-Hématologie, Gustave Roussy, Villejuif, France
| | - Caroline Marty
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| | - Cécile Le Sueur
- Department of Biosciences and Engineering, ETH Zurich, Basel, Switzerland
| | | | - Gaëlle Vertenoeil
- Ludwig Institute for Cancer Research and Université Catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Mira El-Khoury
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| | - Cyril Catelain
- UMS AMMICa-Plateforme Imagerie et Cytométries, Gustave Roussy, Villejuif, France
| | - Philippe Rameau
- UMS AMMICa-Plateforme Imagerie et Cytométries, Gustave Roussy, Villejuif, France
| | - Cyril Gella
- Laboratoire d'Immuno-Hématologie, Gustave Roussy, Villejuif, France
| | | | - Nicole Casadevall
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Assistance Publique des Hôpitaux de Paris, Laboratoire d'Hématologie, Hôpital Saint-Antoine, Paris, France
| | - Rémi Favier
- Assistance Publique des Hôpitaux de Paris, Service d'Hématologie Biologique, Hôpital d'Enfants Armand-Trousseau, Paris, France
| | - Eric Solary
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Département d'Hématologie, Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Bruno Cassinat
- Université de Paris, INSERM UMR-S 1131, Institut de Recherche Saint-Louis (IRSL), Hôpital Saint-Louis, Paris, France
- Assistance Publique des Hôpitaux de Paris, Laboratoire de Biologie Cellulaire
| | - Jean-Jacques Kiladjian
- Université de Paris, INSERM UMR-S 1131, Institut de Recherche Saint-Louis (IRSL), Hôpital Saint-Louis, Paris, France
- Assistance Publique des Hôpitaux de Paris, Centre d'Investigations Cliniques, Hôpital Saint-Louis, Paris, France
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research and Université Catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Florence Pasquier
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Département d'Hématologie, Gustave Roussy, Villejuif, France
| | - Michael E Hochberg
- Institut des Sciences de l'Evolution, University of Montpellier, Montpellier, France
- Santa Fe Institute, Santa Fe, NM
| | - Hana Raslova
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
| | - Jean-Luc Villeval
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
| | - François Girodon
- Laboratoire d'Hématologie, CHU Dijon, Dijon, France
- INSERM, UMR 866, Centre de Recherche, Dijon, France; and
| | - William Vainchenker
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire d'Excellence GR-Ex, Paris, France
- Assistance Publique des Hôpitaux de Paris, Service d'Immunopathologie Clinique, Polyclinique d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - Paul-Henry Cournède
- Université Paris-Saclay, CentraleSupélec, Laboratory MICS (Laboratory of Applied Mathematics and Computer Science), Gif-sur-Yvette, France
| | - Isabelle Plo
- INSERM, Unité Mixte de Recherche (UMR) 1287, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| |
Collapse
|
17
|
Olschok K, Han L, de Toledo MAS, Böhnke J, Graßhoff M, Costa IG, Theocharides A, Maurer A, Schüler HM, Buhl EM, Pannen K, Baumeister J, Kalmer M, Gupta S, Boor P, Gezer D, Brümmendorf TH, Zenke M, Chatain N, Koschmieder S. CALR frameshift mutations in MPN patient-derived iPSCs accelerate maturation of megakaryocytes. Stem Cell Reports 2021; 16:2768-2783. [PMID: 34678208 PMCID: PMC8581168 DOI: 10.1016/j.stemcr.2021.09.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
Calreticulin (CALR) mutations are driver mutations in myeloproliferative neoplasms (MPNs), leading to activation of the thrombopoietin receptor and causing abnormal megakaryopoiesis. Here, we generated patient-derived CALRins5- or CALRdel52-positive induced pluripotent stem cells (iPSCs) to establish an MPN disease model for molecular and mechanistic studies. We demonstrated myeloperoxidase deficiency in granulocytic cells derived from homozygous CALR mutant iPSCs, rescued by repairing the mutation using CRISPR/Cas9. iPSC-derived megakaryocytes showed characteristics of primary megakaryocytes such as formation of demarcation membrane system and cytoplasmic pro-platelet protrusions. Importantly, CALR mutations led to enhanced megakaryopoiesis and accelerated megakaryocytic development in a thrombopoietin-independent manner. Mechanistically, our study identified differentially regulated pathways in mutated versus unmutated megakaryocytes, such as hypoxia signaling, which represents a potential target for therapeutic intervention. Altogether, we demonstrate key aspects of mutated CALR-driven pathogenesis dependent on its zygosity, and found novel therapeutic targets, making our model a valuable tool for clinical drug screening in MPNs. CALR-mutated iPSCs allow efficient modeling of human MPN disease CRISPR-mediated repair of CALR mutations rescues normal iPSC function Megakaryopoiesis in CALR-mutated iPSCs is hyperplastic and accelerated Transcriptome screen of mutated megakaryocytes identifies novel therapeutic options
Collapse
Affiliation(s)
- Kathrin Olschok
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Lijuan Han
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany; Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Marcelo A S de Toledo
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Janik Böhnke
- Institute for Biomedical Engineering, Department of Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Martin Graßhoff
- Institute for Computational Genomics Joint Research Center for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Ivan G Costa
- Institute for Computational Genomics Joint Research Center for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Alexandre Theocharides
- Division of Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Angela Maurer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Herdit M Schüler
- Institute for Human Genetics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Eva Miriam Buhl
- Institute for Pathology, Electron Microscopy Facility, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Kristina Pannen
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Julian Baumeister
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Milena Kalmer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Siddharth Gupta
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Peter Boor
- Institute for Pathology, Electron Microscopy Facility, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Deniz Gezer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Martin Zenke
- Institute for Biomedical Engineering, Department of Cell Biology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany.
| |
Collapse
|
18
|
Loscocco GG, Guglielmelli P, Gangat N, Rossi E, Mannarelli C, Betti S, Maccari C, Ramundo F, Jadoon Y, Gesullo F, Ceglie S, Paoli C, Pardanani A, De Stefano V, Tefferi A, Vannucchi AM. Clinical and molecular predictors of fibrotic progression in essential thrombocythemia: A multicenter study involving 1607 patients. Am J Hematol 2021; 96:1472-1480. [PMID: 34424575 PMCID: PMC9293196 DOI: 10.1002/ajh.26332] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 12/31/2022]
Abstract
The current retrospective study involving a total of 1607 patients was designed to identify clinical and molecular variables that were predictive of inferior myelofibrosis‐free survival (MFS) in WHO‐defined essential thrombocythemia (ET), utilizing three independent patient cohorts: University of Florence, Italy (n = 718); Mayo Clinic, USA (n = 479) and Policlinico Gemelli, Catholic University, Rome, Italy (n = 410). The Florence patient cohort was first examined to identify independent risk factors for MFS, which included age > 60 years (HR 2.5, 95% CI 1.3–4.9), male sex (2.1, 1.2–3.9), palpable splenomegaly (2.1, 1.2–3.9), CALR 1/1‐like or MPL mutation (3.4, 1.9–6.1) and JAK2V617F variant allele frequency > 35% (4.2, 1.6–10.8). Subsequently, an operational molecular risk category was developed and validated in the other two cohorts from Mayo Clinic and Rome: “high molecular risk” category included patients with JAK2V617F VAF >35%, CALR type 1/1‐like or MPL mutations; all other driver mutation profiles were assigned to “low molecular risk” category. The former, compared to the latter molecular risk category, displayed significantly higher risk of fibrotic transformation: Florence cohort with respective fibrotic transformation risk rates of 8% vs. 1.2% at 10 years and 33% vs. 8% at 20 years (p < 0.001; HR 6.1; 95% CI 3.2–11.7); Mayo Cohort, 16% vs. 7% at 10 years and 44% vs. 25% at 20 years (p < 0.001; HR 2.5; 95% CI 1.6–4.1); and Rome cohort 7.8% vs. 4.6% at 10 years and 31.2% vs. 7.1% at 20 years (p = 0.007, HR 2.7; 95% CI 1.3–5.8). The present study provides practically useful risk signals for fibrotic transformation in ET and facilitates identification of patients who require close monitoring and appropriate counseling.
Collapse
Affiliation(s)
- Giuseppe G. Loscocco
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | - Naseema Gangat
- Divisions of Hematology Mayo Clinic Rochester Minnesota USA
| | - Elena Rossi
- Section of Hematology, Department of Radiological and Hematological Sciences Catholic University Rome Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS Rome Italy
| | - Carmela Mannarelli
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | - Silvia Betti
- Fondazione Policlinico Universitario A. Gemelli IRCCS Rome Italy
| | - Chiara Maccari
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | - Francesco Ramundo
- Section of Hematology, Department of Radiological and Hematological Sciences Catholic University Rome Italy
| | - Yamna Jadoon
- Divisions of Hematology Mayo Clinic Rochester Minnesota USA
| | - Francesca Gesullo
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | - Sara Ceglie
- Section of Hematology, Department of Radiological and Hematological Sciences Catholic University Rome Italy
| | - Chiara Paoli
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| | | | - Valerio De Stefano
- Section of Hematology, Department of Radiological and Hematological Sciences Catholic University Rome Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS Rome Italy
| | - Ayalew Tefferi
- Divisions of Hematology Mayo Clinic Rochester Minnesota USA
| | - Alessandro M. Vannucchi
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐Universitaria Careggi University of Florence Florence Italy
| |
Collapse
|
19
|
Penna D. New Horizons in Myeloproliferative Neoplasms Treatment: A Review of Current and Future Therapeutic Options. Medicina (B Aires) 2021; 57:medicina57111181. [PMID: 34833399 PMCID: PMC8619471 DOI: 10.3390/medicina57111181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 11/29/2022] Open
Abstract
Philadelphia-negative myeloproliferative neoplasms (MPN) are aggressive diseases characterized by clonal proliferation of myeloid stem cells. The clonal process leads to excessive red cells production, platelets production, and bone marrow fibrosis. According to the phenotype, MPN can be classified as polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). MPN patients have shortened survival due to the increased risk of thrombosis, hemorrhages, and transformation to acute myeloid leukemia (AML). Prognosis is variable, with a shorter life expectancy in myelofibrosis. Currently, drug therapy can reduce symptoms, splenomegaly, and risk of thrombosis. Still, some patients can be resistant or intolerant to the treatment. At the same time, allogeneic stem cell transplant (ASCT) is the only treatment modality with the potential to cure the disease. Nevertheless, the ASCT is reserved for high-risk leukemic progression patients due to the risk of treatment-related death and comorbidity. Therefore, there is a need for new drugs that can eradicate clonal hematopoiesis and prevent progression to more aggressive myeloid neoplasms. Thanks to the better understanding of the disease’s molecular pathogenesis, many new potentially disease-modifying drugs have been developed and are currently in clinical trials. This review explores the most promising new drugs currently in clinical trials.
Collapse
Affiliation(s)
- Domenico Penna
- Hematology Unit, Azienda Unità Sanitaria Locale—IRCCS, 42123 Reggio Emilia, Italy; ; Tel.: +39-522-296-623
- Ph.D. Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 42121 Modena, Italy
| |
Collapse
|
20
|
Shide K. Calreticulin mutations in myeloproliferative neoplasms. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 365:179-226. [PMID: 34756244 DOI: 10.1016/bs.ircmb.2021.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Calreticulin (CALR) is a chaperone present in the endoplasmic reticulum, which is involved in the quality control of N-glycosylated proteins and storage of calcium ions. In 2013, the C-terminal mutation in CALR was identified in half of the patients with essential thrombocythemia and primary myelofibrosis who did not have a JAK2 or MPL mutation. The results of 8 years of intensive research are changing the clinical practice associated with treating myeloproliferative neoplasms (MPNs). The presence or absence of CALR mutations and their mutation types already provide important information for diagnosis and treatment decision making. In addition, the interaction with the thrombopoietin receptor MPL, which is the main mechanism of transformation by CALR mutation, and the expression of the mutant protein on the cell surface have a great potential as targets for molecular-targeted drugs and immunotherapy. This chapter presents recent findings on the clinical significance of the CALR mutation and the molecular basis by which this mutation drives MPNs.
Collapse
Affiliation(s)
- Kotaro Shide
- Division of Haematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
| |
Collapse
|
21
|
Greenfield G, McMullin MF, Mills K. Molecular pathogenesis of the myeloproliferative neoplasms. J Hematol Oncol 2021; 14:103. [PMID: 34193229 PMCID: PMC8246678 DOI: 10.1186/s13045-021-01116-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
The Philadelphia negative myeloproliferative neoplasms (MPN) compromise a heterogeneous group of clonal myeloid stem cell disorders comprising polycythaemia vera, essential thrombocythaemia and primary myelofibrosis. Despite distinct clinical entities, these disorders are linked by morphological similarities and propensity to thrombotic complications and leukaemic transformation. Current therapeutic options are limited in disease-modifying activity with a focus on the prevention of thrombus formation. Constitutive activation of the JAK/STAT signalling pathway is a hallmark of pathogenesis across the disease spectrum with driving mutations in JAK2, CALR and MPL identified in the majority of patients. Co-occurring somatic mutations in genes associated with epigenetic regulation, transcriptional control and splicing of RNA are variably but recurrently identified across the MPN disease spectrum, whilst epigenetic contributors to disease are increasingly recognised. The prognostic implications of one MPN diagnosis may significantly limit life expectancy, whilst another may have limited impact depending on the disease phenotype, genotype and other external factors. The genetic and clinical similarities and differences in these disorders have provided a unique opportunity to understand the relative contributions to MPN, myeloid and cancer biology generally from specific genetic and epigenetic changes. This review provides a comprehensive overview of the molecular pathophysiology of MPN exploring the role of driver mutations, co-occurring mutations, dysregulation of intrinsic cell signalling, epigenetic regulation and genetic predisposing factors highlighting important areas for future consideration.
Collapse
Affiliation(s)
- Graeme Greenfield
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK.
| | | | - Ken Mills
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| |
Collapse
|
22
|
Induced Pluripotent Stem Cells Enable Disease Modeling and Drug Screening in Calreticulin del52 and ins5 Myeloproliferative Neoplasms. Hemasphere 2021; 5:e593. [PMID: 34131633 PMCID: PMC8196125 DOI: 10.1097/hs9.0000000000000593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Mutations in the calreticulin (CALR) gene are seen in about 30% of essential thrombocythemia and primary myelofibrosis patients. To address the contribution of the human CALR mutants to the pathogenesis of myeloproliferative neoplasms (MPNs) in an endogenous context, we modeled the CALRdel52 and CALRins5 mutants by induced pluripotent stem cell (iPSC) technology using CD34+ progenitors from 4 patients. We describe here the generation of several clones of iPSC carrying heterozygous CALRdel52 or CALRins5 mutations. We showed that CALRdel52 induces a stronger increase in progenitors than CALRins5 and that both CALRdel52 and CALRins5 mutants favor an expansion of the megakaryocytic lineage. Moreover, we found that both CALRdel52 and CALRins5 mutants rendered colony forming unit–megakaryocyte (CFU-MK) independent from thrombopoietin (TPO), and promoted a mild constitutive activation level of signal transducer and activator of transcription 3 in megakaryocytes. Unexpectedly, a mild increase in the sensitivity of colony forming unit-granulocyte (CFU-G) to granulocyte-colony stimulating factor was also observed in iPSC CALRdel52 and CALRins5 compared with control iPSC. Moreover, CALRdel52-induced megakaryocytic spontaneous growth is more dependent on Janus kinase 2/phosphoinositide 3-kinase/extracellular signal-regulated kinase than TPO-mediated growth and opens a therapeutic window for treatments in CALR-mutated MPN. The iPSC models described here represent an interesting platform for testing newly developed inhibitors. Altogether, this study shows that CALR-mutated iPSC recapitulate MPN phenotypes in vitro and may be used for drug screening.
Collapse
|
23
|
Lambert J, Saliba J, Calderon C, Sii-Felice K, Salma M, Edmond V, Alvarez JC, Delord M, Marty C, Plo I, Kiladjian JJ, Soler E, Vainchenker W, Villeval JL, Rousselot P, Prost S. PPARγ agonists promote the resolution of myelofibrosis in preclinical models. J Clin Invest 2021; 131:136713. [PMID: 33914703 DOI: 10.1172/jci136713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Myelofibrosis (MF) is a non-BCR-ABL myeloproliferative neoplasm associated with poor outcomes. Current treatment has little effect on the natural history of the disease. MF results from complex interactions between (a) the malignant clone, (b) an inflammatory context, and (c) remodeling of the bone marrow (BM) microenvironment. Each of these points is a potential target of PPARγ activation. Here, we demonstrated the therapeutic potential of PPARγ agonists in resolving MF in 3 mouse models. We showed that PPARγ agonists reduce myeloproliferation, modulate inflammation, and protect the BM stroma in vitro and ex vivo. Activation of PPARγ constitutes a relevant therapeutic target in MF, and our data support the possibility of using PPARγ agonists in clinical practice.
Collapse
Affiliation(s)
- Juliette Lambert
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Department of Hematology and Oncology, Centre Hospitalier de Versailles, Le Chesnay, France.,Opale Carnot Institute, Paris, France
| | - Joseph Saliba
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Carolina Calderon
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Opale Carnot Institute, Paris, France
| | - Karine Sii-Felice
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Mohammad Salma
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | - Valérie Edmond
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Jean-Claude Alvarez
- Département de Pharmacologie-Toxicologie, Hôpitaux Universitaires Paris Ile-de-France Ouest, AP-HP, Hôpital Raymond-Poincaré, FHU Sepsis, Garches, France.,MasSpecLab, Plateforme de spectrométrie de masse, INSERM U-1173, Université Paris-Saclay (Versailles Saint-Quentin-en-Yvelines), UFR des sciences de la santé, Montigny-le-Bretonneux, France
| | - Marc Delord
- Recherche Clinique, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Caroline Marty
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Jean-Jacques Kiladjian
- Opale Carnot Institute, Paris, France.,Université de Paris, AP-HP, Hôpital Saint-Louis, Centre d'Investigations Cliniques CIC 1427, INSERM, Paris, France
| | - Eric Soler
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | | | - Jean-Luc Villeval
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Philippe Rousselot
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Department of Hematology and Oncology, Centre Hospitalier de Versailles, Le Chesnay, France.,Opale Carnot Institute, Paris, France.,Université Paris-Saclay (Versailles Saint-Quentin-en-Yvelines), UFR des sciences de la santé, Montigny-le-Bretonneux, France
| | - Stéphane Prost
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Opale Carnot Institute, Paris, France
| |
Collapse
|
24
|
Achyutuni S, Nivarthi H, Majoros A, Hug E, Schueller C, Jia R, Varga C, Schuster M, Senekowitsch M, Tsiantoulas D, Kavirayani A, Binder CJ, Bock C, Zagrijtschuk O, Kralovics R. Hematopoietic expression of a chimeric murine-human CALR oncoprotein allows the assessment of anti-CALR antibody immunotherapies in vivo. Am J Hematol 2021; 96:698-707. [PMID: 33761144 DOI: 10.1002/ajh.26171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/18/2021] [Accepted: 03/21/2021] [Indexed: 12/30/2022]
Abstract
Myeloproliferative neoplasms (MPNs) are characterized by a pathologic expansion of myeloid lineages. Mutations in JAK2, CALR and MPL genes are known to be three prominent MPN disease drivers. Mutant CALR (mutCALR) is an oncoprotein that interacts with and activates the thrombopoietin receptor (MPL) and represents an attractive target for targeted therapy of CALR mutated MPN. We generated a transgenic murine model with conditional expression of the human mutant exon 9 (del52) from the murine endogenous Calr locus. These mice develop essential thrombocythemia like phenotype with marked thrombocytosis and megakaryocytosis. The disease exacerbates with age showing prominent signs of splenomegaly and anemia. The disease is transplantable and mutCALR stem cells show proliferative advantage when compared to wild type stem cells. Transcriptome profiling of hematopoietic stem cells revealed oncogenic and inflammatory gene expression signatures. To demonstrate the applicability of the transgenic animals for immunotherapy, we treated mice with monoclonal antibody raised against the human mutCALR. The antibody treatment lowered platelet and stem cell counts in mutant mice. Secretion of mutCALR did not constitute a significant antibody sink. This animal model not only recapitulates human MPN but also serves as a relevant model for testing immunotherapeutic strategies targeting epitopes of the human mutCALR.
Collapse
Affiliation(s)
- Sarada Achyutuni
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Harini Nivarthi
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Eva Hug
- MyeloPro Diagnostics and Research GmbH, Vienna, Austria
| | - Christina Schueller
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ruochen Jia
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- MyeloPro Diagnostics and Research GmbH, Vienna, Austria
| | - Cecilia Varga
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Schuster
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Martin Senekowitsch
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Dimitris Tsiantoulas
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph Bock
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Robert Kralovics
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| |
Collapse
|
25
|
Activated IL-6 signaling contributes to the pathogenesis of, and is a novel therapeutic target for, CALR-mutated MPNs. Blood Adv 2021; 5:2184-2195. [PMID: 33890979 DOI: 10.1182/bloodadvances.2020003291] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/14/2021] [Indexed: 02/08/2023] Open
Abstract
Calreticulin (CALR), an endoplasmic reticulum-associated chaperone, is frequently mutated in myeloproliferative neoplasms (MPNs). Mutated CALR promotes downstream JAK2/STAT5 signaling through interaction with, and activation of, the thrombopoietin receptor (MPL). Here, we provide evidence of a novel mechanism contributing to CALR-mutated MPNs, represented by abnormal activation of the interleukin 6 (IL-6)-signaling pathway. We found that UT7 and UT7/mpl cells, engineered by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) to express the CALR type 1-like (DEL) mutation, acquired cytokine independence and were primed to the megakaryocyte (Mk) lineage. Levels of IL-6 messenger RNA (mRNA), extracellular-released IL-6, membrane-associated glycoprotein 130 (gp130), and IL-6 receptor (IL-6R), phosphorylated JAK1 and STAT3 (p-JAK1 and p-STAT3), and IL-6 promoter region occupancy by STAT3 all resulted in increased CALR DEL cells in the absence of MPL stimulation. Wild-type, but not mutated, CALR physically interacted with gp130 and IL-6R, downregulating their expression on the cell membrane. Agents targeting gp130 (SC-144), IL-6R (tocilizumab [TCZ]), and cell-released IL-6 reduced proliferation of CALR DEL as well as CALR knockout cells, supporting a mutated CALR loss-of-function model. CD34+ cells from CALR-mutated patients showed increased levels of IL-6 mRNA and p-STAT3, and colony-forming unit-Mk growth was inhibited by either SC144 or TCZ, as well as an IL-6 antibody, supporting cell-autonomous activation of the IL-6 pathway. Targeting IL-6 signaling also reduced colony formation by CD34+ cells of JAK2V617F-mutated patients. The combination of TCZ and ruxolitinib was synergistic at very low nanomolar concentrations. Overall, our results suggest that target inhibition of IL-6 signaling may have therapeutic potential in CALR, and possibly JAK2V617F, mutated MPNs.
Collapse
|
26
|
Benlabiod C, Dagher T, Marty C, Villeval JL. Lessons from mouse models of MPN. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 366:125-185. [PMID: 35153003 DOI: 10.1016/bs.ircmb.2021.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Over the past decades, a variety of MPN mouse models have been developed to express in HSC the main mutations identified in patients: JAK2V617F, CALRdel52 or ins5 and MPLW515L. These models mimic quite faithfully human PV or ET with their natural evolutions into MF and their hemostasis complications, demonstrating the driver function of these mutations in MPN. Here, we review these models and show how they have improved our general understanding of MPN regarding (1) the mechanisms of fibrosis, thrombosis/hemorrhages and disease initiation, (2) the roles of additional mutations and signaling pathways in disease progression and (3) the preclinical development of novel therapies. We also address controversial results between these models and remind how these models may differ from human MPN onset and also how basically mice are not humans, encouraging caution when one draw lessons from mice to humans. Furthermore, the contribution of germline genetic predisposition, HSC and niche aging, metabolic, oxidative, replicative or genotoxic stress, inflammation, immune escape and additional mutations need to be considered in further investigations to encompass the full complexity of human MPN in mice.
Collapse
Affiliation(s)
- Camelia Benlabiod
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France
| | - Tracy Dagher
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France
| | - Caroline Marty
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France.
| | - Jean-Luc Villeval
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France.
| |
Collapse
|
27
|
The Contemporary Approach to CALR-Positive Myeloproliferative Neoplasms. Int J Mol Sci 2021; 22:ijms22073371. [PMID: 33806036 PMCID: PMC8038093 DOI: 10.3390/ijms22073371] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 12/20/2022] Open
Abstract
CALR mutations are a revolutionary discovery and represent an important hallmark of myeloproliferative neoplasms (MPN), especially essential thrombocythemia and primary myelofibrosis. To date, several CALR mutations were identified, with only frameshift mutations linked to the diseased phenotype. It is of diagnostic and prognostic importance to properly define the type of CALR mutation and subclassify it according to its structural similarities to the classical mutations, a 52-bp deletion (type 1 mutation) and a 5-bp insertion (type 2 mutation), using a statistical approximation algorithm (AGADIR). Today, the knowledge on the pathogenesis of CALR-positive MPN is expanding and several cellular mechanisms have been recognized that finally cause a clonal hematopoietic expansion. In this review, we discuss the current basis of the cellular effects of CALR mutants and the understanding of its implementation in the current diagnostic laboratorial and medical practice. Different methods of CALR detection are explained and a diagnostic algorithm is shown that aids in the approach to CALR-positive MPN. Finally, contemporary methods joining artificial intelligence in accordance with molecular-genetic biomarkers in the approach to MPN are presented.
Collapse
|
28
|
Stetka J, Skoda RC. Mouse models of myeloproliferative neoplasms for pre-clinical testing of novel therapeutic agents. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2021; 165:26-33. [PMID: 33542546 DOI: 10.5507/bp.2021.004] [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: 12/14/2020] [Accepted: 01/08/2021] [Indexed: 11/23/2022] Open
Abstract
Myeloproliferative neoplasms (MPN), are clonal hematopoietic stem cell (HSC) disorders driven by gain-of-function mutations in JAK2 (JAK2-V617F), CALR or MPL genes. MPN treatment options currently mainly consist of cytoreductive therapy with hydroxyurea and JAK2 inhibitors such as ruxolitinib and fedratinib. Pegylated interferon-alpha can induce complete molecular remission (CMR) in some MPN patients when applied at early stages of disease. The ultimate goal of modern MPN treatment is to develop novel therapies that specifically target mutant HSCs in MPN and consistently induce CMR. Basic research has identified a growing number of candidate drugs with promising effects in vitro. A first step on the way to developing these compounds into drugs approved for treatment of MPN patients often consists of examining the effects in vivo using pre-clinical mouse models of MPN. Here we review the current state of MPN mouse models and the experimental setup for their optimal use in drug testing. In addition to novel compounds, combinatorial therapeutic approaches are often considered for the treatment of MPN. Optimized and validated mouse models can provide an efficient way to rapidly assess and select the most promising combinations and thereby contribute to accelerating the development of novel therapies of MPN.
Collapse
Affiliation(s)
- Jan Stetka
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, Switzerland.,Department of Biology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Radek C Skoda
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|