1
|
Loscocco GG, Guglielmelli P. Targeted Therapies in Myelofibrosis: Present Landscape, Ongoing Studies, and Future Perspectives. Am J Hematol 2025; 100 Suppl 4:30-50. [PMID: 40062529 PMCID: PMC12067168 DOI: 10.1002/ajh.27658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/20/2024] [Accepted: 02/27/2025] [Indexed: 05/13/2025]
Abstract
Myelofibrosis (MF) is a myeloproliferative neoplasm that is accompanied by driver JAK2, CALR, or MPL mutations in more than 90% of cases, leading to constitutive activation of the JAK-STAT pathway. MF is a multifaceted disease characterized by trilineage myeloid proliferation with prominent megakaryocyte atypia and bone marrow fibrosis, as well as splenomegaly, constitutional symptoms, ineffective erythropoiesis, extramedullary hematopoiesis, and a risk of leukemic progression and shortened survival. Therapy can range from observation alone in lower-risk and asymptomatic patients to allogeneic hematopoietic stem cell transplantation, which is the only potentially curative treatment capable of prolonging survival, although burdened by significant morbidity and mortality. The discovery of the JAK2 V617F mutation prompted the development of JAK inhibitors (JAKi) including the first-in-class JAK1/JAK2 inhibitor ruxolitinib and subsequent approval of fedratinib, pacritinib, and momelotinib. The latter has shown erythropoietic benefits by suppressing hepcidin expression via activin A receptor type 1 (ACVR1) inhibition, as well as reducing splenomegaly and symptoms. However, the current JAKi behave as anti-inflammatory drugs without a major impact on survival or disease progression. A better understanding of the genetics, mechanisms of fibrosis, cytopenia, and the role of inflammatory cytokines has led to the development of numerous therapeutic agents that target epigenetic regulation, signaling, telomerase, cell cycle, and apoptosis, nuclear export, and pro-fibrotic cytokines. Selective JAK2 V617F inhibitors and targeting of mutant CALR by immunotherapy are the most intriguing and promising approaches. This review focuses on approved and experimental treatments for MF, highlighting their biological background.
Collapse
Affiliation(s)
- Giuseppe G. Loscocco
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐ Universitaria CareggiUniversity of FlorenceFlorenceItaly
- Division of HematologyMayo ClinicRochesterMinnesotaUSA
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero‐ Universitaria CareggiUniversity of FlorenceFlorenceItaly
| |
Collapse
|
2
|
Naseer S, Singh A, Shrivastva S, Singh RK, Chowdhury S, Dey CS, Roy A. Differential regulation of mTORC2 signalling by type I and type II calreticulin (CALR) driver mutations of myeloproliferative neoplasm. Cell Commun Signal 2025; 23:221. [PMID: 40355863 PMCID: PMC12067760 DOI: 10.1186/s12964-025-02212-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Calreticulin (CALR) is an endoplasmic reticulum chaperone. Frameshift mutations in CALR were discovered in patients with myeloproliferative neoplasm showing increased platelet counts. The frameshift was observed in the last exon of CALR, leading to a novel C-terminal tail. Calreticulin mutations were categorised into Type I and Type II depending upon the extent of retention of CALR WT sequences. Clinically, Type I mutations induced myelofibrosis, while Type II mutations were associated with early onset of the disease. Both mutations induced ligand-independent activation of the thrombopoietin receptor (TpoR) and consequently enhanced platelet production. However, no specific difference in signalling mechanism could be demonstrated between them. Using over-expression of CALR WT, CALR ∆52 (Type I) and CALR ins5 (Type II) in HEK cells, we showed that Type I CALR mutations downregulated the basal mTORC2 signalling without affecting mTORC1. The decrease in basal mTORC2 signalling was attributed to CALR ∆52-induced increased expression of c-JUN through occupation of the enhancer sequences of jun. Furthermore, increased c-JUN expression decreased the expression of RICTOR, a component of mTORC2. Strikingly, overexpression of RICTOR or knockdown of c-JUN reversed the inhibitory effect of CALR ∆52 on mTORC2 activity. Finally, we demonstrated that CALR ∆52 decreased the glucose uptake and cellular ATP levels in a c-JUN-mTORC2-dependent manner. These findings not only contribute to our understanding of the molecular mechanisms underlying mutant CALR driven myeloproliferative neoplasm but also provide potential therapeutic targets against the disease.
Collapse
Affiliation(s)
- Saadia Naseer
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Aditi Singh
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Saurabh Shrivastva
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Rishi Kant Singh
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Shayeri Chowdhury
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Anita Roy
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
3
|
Beauverd Y, Ianotto JC, Thaw KH, Sobas M, Sadjadian P, Curto-Garcia N, Shih LY, Devos T, Krochmalczyk D, Galli S, Bieniaszewska M, Seferynska I, McMullin MF, Armatys A, Spalek A, Waclaw J, Zdrenghea MT, Legros L, Girodon F, Lewandowski K, Bellosillo B, Samuelsson J, Abuin Blanco A, Cony-Makhoul P, Collins A, James C, Kusec R, Lauermannova M, Noya MS, Skowronek M, Szukalski L, Szmigielska-Kaplon A, Wondergem M, Dudchenko I, Gora-Tybor J, Laribi K, Kulikowska de Nałęcz A, Demory JL, Le Dû K, Zweegman S, Besses Raebel C, Skoda RC, Giraudier S, Griesshammer M, Kiladjian JJ, Harrison CN. Impact of treatment for adolescent and young adults with essential thrombocythemia and polycythemia vera. Leukemia 2025; 39:1135-1145. [PMID: 40074852 PMCID: PMC12055580 DOI: 10.1038/s41375-025-02545-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/11/2024] [Accepted: 02/18/2025] [Indexed: 03/14/2025]
Abstract
Essential thrombocythemia (ET) and polycythemia vera (PV) are rare in adolescent and young adult (AYA). These conditions, similar to those in older patients, are linked with thrombotic complications and the potential progression to secondary myelofibrosis (sMF). This retrospective study of ET and PV patients diagnosed before age 25 evaluated complication rates and impact of cytoreductive drugs on outcomes. Among 348 patients (278 ET, 70 PV) with a median age of 20 years, the of thrombotic events was 1.9 per 100 patient-years. Risk factors for thrombosis included elevated white blood cell count (>11 × 109/L) (HR: 2.7, p = 0.012) and absence of splenomegaly at diagnosis (HR: 5.7, p = 0.026), while cytoreductive drugs did not reduce this risk. The incidence of sMF was 0.7 per 100 patient-years. CALR mutation (HR: 6.0, p < 0.001) and a history of thrombosis (HR: 3.8, p = 0.015) were associated with sMF risk. Interferon as a first-line treatment significantly improved myelofibrosis-free survival compared to other treatments or the absence of cytoreduction (p = 0.046). Although cytoreduction did not affect thrombotic event, early interferon use reduced sMF risk. These findings support interferon use to mitigate sMF risk in AYA ET and PV patients.
Collapse
Affiliation(s)
- Yan Beauverd
- Hematology Division and Faculty of Medicine, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | | | - Kyaw Htin Thaw
- Haematology Department, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | | | - Parvis Sadjadian
- University Clinic for Hematology, Oncology, Hemostaseology and Palliative Care, Johannes Wesling Medical Center, University of Bochum, Minden, Germany
| | - Natalia Curto-Garcia
- Haematology Department, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Lee-Yung Shih
- Division of Hematology-Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Timothy Devos
- Department of Hematology, University Hospitals Leuven and Department of Microbiology and Immunology, Laboratory of Molecular Immunology (Rega Institute), KU Leuven, Leuven, Belgium
| | - Dorota Krochmalczyk
- Department of Hematology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Serena Galli
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Maria Bieniaszewska
- Hematology and Transplantation Department, Medical University and Clinical Center, Gdansk, Poland
| | - Ilona Seferynska
- Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Mary Frances McMullin
- Haematology, Belfast City Hospital, Queen's University Belfast, Belfast, United Kingdom
| | - Anna Armatys
- Hematology Department, Jagiellonian University Hospital, Krakow, Poland
| | - Adrianna Spalek
- Hematology Department, Jagiellonian University Hospital, Krakow, Poland
| | - Joanna Waclaw
- Department of Hematology, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Mihnea Tudor Zdrenghea
- Iuliu Hatieganu University of Medicine and Pharmacy, Department of Hematology, Cluj-Napoca, Romania
| | - Laurence Legros
- Hematology Department, AP-HP, University of Paris Saclay, Bicêtre Hospital, Paris, France
| | - Francois Girodon
- Laboratory of Biological Hematology, University Hospital, Dijon, France
| | - Krzysztof Lewandowski
- Hematology and Bone Marrow Transplantation Department, University of Medical Sciences, Poznan, Poland
| | - Beatriz Bellosillo
- Hematology Department, Hospital del Mar, Hospital del Mar Research Institute, Barcelona, Spain
| | - Jan Samuelsson
- Hematology Department, University Hospital, Linkoping, Sweden
| | - Aitor Abuin Blanco
- Servicio de Hematología. Hospital Universitario Lucus Augusti, Lugo, Spain
| | | | - Angela Collins
- Department of Haematology, Norfolk and Norwich University Hospitals National Health Service Trust, Norwich, United Kingdom
| | - Chloe James
- Biology of Cardiovascular Diseases, University of Bordeaux, INSERM, UMR1034, Pessac, France
| | - Rajko Kusec
- Department of Hematology, University Hospital Dubrava, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Marie Lauermannova
- Institute of Hematology and Blood Transfusion, Prague, Prague, Czech Republic
| | | | | | - Lukasz Szukalski
- Department of Hematology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Torun, Poland
| | | | - Marielle Wondergem
- Department of Hematology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Iryna Dudchenko
- Department of Internal Medicine with Respiratory Medicine Center, Academic and Research Medical institute, Sumy State University, Sumy, Ukraine
| | | | - Kamel Laribi
- Hematology Department, Le Mans Hospital, Le Mans, France
| | | | - Jean-Loup Demory
- Department of Hematology, St. Vincent De Paul Hospital, Lille, France
| | | | - Sonja Zweegman
- Department of Hematology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Carlos Besses Raebel
- Hematology Department, Hospital del Mar, Hospital del Mar Research Institute, Barcelona, Spain
| | - Radek C Skoda
- Experimental Hematology, Department of Biomedicine, University Hospital and University, Basel, Switzerland
| | - Stephane Giraudier
- Cellular Biology Department, INSERM UMRS 1131, St Louis Hospital, APHP, Paris, France
| | - Martin Griesshammer
- University Clinic for Hematology, Oncology, Hemostaseology and Palliative Care, Johannes Wesling Medical Center, University of Bochum, Minden, Germany
| | | | - Claire N Harrison
- Guy's and St. Thomas' NHS Foundation Trust, London, ENG, United Kingdom
| |
Collapse
|
4
|
Coltoff A, Mascarenhas J. Fedratinib in 2025 and beyond: indications and future applications. Blood Adv 2025; 9:1907-1915. [PMID: 39951613 PMCID: PMC12008686 DOI: 10.1182/bloodadvances.2024015365] [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: 11/14/2024] [Revised: 01/13/2025] [Accepted: 02/08/2025] [Indexed: 02/16/2025] Open
Abstract
ABSTRACT Dysregulated JAK/STAT signaling underlies the pathogenesis of myelofibrosis, a myeloproliferative neoplasm characterized by cytopenias, splenomegaly, and constitutional symptoms. JAK inhibitors, such as fedratinib, are the primary therapeutic option for patients with high-risk or symptomatic myelofibrosis. Fedratinib has characteristics that distinguish it from other commercially available JAK inhibitors, such as its preferential inhibition of JAK2 and its inhibitory effects on kinases such as Fms-like tyrosine kinase 3 and BRD4. Fedratinib is most often used in the second-line setting after intolerance or resistance to other JAK inhibitors, but there is substantial evidence that it is an effective first-line option in the appropriate patient population. Prevention and early treatment of fedratinib-related gastrointestinal toxicity is key to maintaining adequate drug exposure, and clinicians must remain vigilant for Wernicke encephalopathy during treatment. Fedratinib's JAK2 selectivity and kinome profile make it an appealing agent for alternative indications, such as myelodysplastic/myeloproliferative neoplasms and maintenance after bone marrow transplantation, which are under active investigation.
Collapse
Affiliation(s)
- Alexander Coltoff
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - John Mascarenhas
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
5
|
Guy A, Morange PE, James C. How I approach the treatment of thrombotic complications in patients with myeloproliferative neoplasms. Blood 2025; 145:1769-1779. [PMID: 39541574 DOI: 10.1182/blood.2024025627] [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: 07/24/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
ABSTRACT Arterial and venous thromboses are the most significant complications in patients with myeloproliferative neoplasms (MPNs), with the primary treatment goal being thrombotic risk reduction. In MPN with no history of thrombosis, primary prevention mainly involves the use of aspirin, and cytoreduction is added in high-risk patients. However, thrombotic complications can unveil an MPN in ∼20% of cases, necessitating the initiation of both antithrombotic therapy for the thrombosis and cytoreductive treatment for the MPN. The duration of anticoagulant therapy after an initial venous thromboembolic event is subject to discussion. Furthermore, the occurrence of a thrombotic complication in patients with a known diagnosis of MPN prompts a reconsideration of both antithrombotic and hematologic management. This review uses case-based discussions to explore the management of thrombotic complications in patients with MPN. It addresses the nature and duration of antithrombotic treatments, as well as the approach to cytoreduction. Special attention is given to the place of direct oral anticoagulants and to the management of patients with MPN with splanchnic vein thrombosis, which is disproportionately common in this group.
Collapse
Affiliation(s)
- Alexandre Guy
- Laboratory of Hematology, University Hospital Bordeaux, Pessac, France
- Biologie des Maladies Cardiovasculaires, U-1034, University of Bordeaux, INSERM, Pessac, France
| | - Pierre-Emmanuel Morange
- Laboratory of Hematology, Assistance Publique-Hôpitaux de Marseille, Marseille, France
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Centre de Recherche en Cardiovasculaire et Nutrition, University of Aix-Marseille, INSERM, Marseille, France
| | - Chloé James
- Laboratory of Hematology, University Hospital Bordeaux, Pessac, France
- Biologie des Maladies Cardiovasculaires, U-1034, University of Bordeaux, INSERM, Pessac, France
| |
Collapse
|
6
|
Liu X, Krishnamurthy K, Naeem R, Goldstein DY. A comparison of sequential polymerase chain reaction-based cascade testing vs next-generation sequencing in molecular profiling of myeloproliferative neoplasms: improving testing strategies in light of evolving molecular landscapes. Lab Med 2025:lmaf004. [PMID: 40238187 DOI: 10.1093/labmed/lmaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
INTRODUCTION Somatic mutations in the JAK2, CALR, and MPL genes are traditionally tested using a cascading reflex algorithm in BCR::ABL1-negative myeloproliferative neoplasms (MPNs). However, next-generation sequencing (NGS) has revealed that these variants may coexist, exposing limitations in current testing practices. METHODS This pilot study analyzed 3 JAK2 p.V617F-positive MPN cases and 3 cases negative for classical driver mutations using the Oncomine Myeloid Assay GX v2 assay (Thermo Fisher Scientific) on the Genexus Integrated Sequencer (Thermo Fisher Scientific). RESULTS JAK2 p.V617F status was 100% concordant between polymerase chain reaction (PCR) and NGS. Next-generation sequencing detected a concurrent driver MPL mutation in 1 case, an SF3B1 variant in 1 case, and an IDH2 variant in a JAK2-positive case that have established prognostic and therapeutic significance. In cases negative for conventional targets, NGS detected DNMT3A and TET2 variants, which are associated with clonal hematopoiesis and MPN initiation. One case had a JAK2 p.V617F alteration at a variant allele frequency of 0.9%, below the NGS-reportable range but detectable by PCR, adding another caveat to profiling of MPNs. DISCUSSION Next-generation sequencing provides comprehensive molecular profiling in patients with MPNs, identifying additional prognostic and therapeutic markers. However, PCR remains superior for detecting low-variant allele frequency variants. We propose an updated MPN testing strategy that integrates PCR and NGS within a reflex algorithm to optimize diagnostics and therapeutic guidance.
Collapse
Affiliation(s)
- Xiaowei Liu
- Department of Pathology, Montefiore Medical Center, Bronx, NY, United States
| | | | - Rizwan Naeem
- Department of Pathology, Montefiore Medical Center, Bronx, NY, United States
| | - D Yitzchak Goldstein
- Department of Pathology, Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
7
|
Semenuk L, Palazzolo A, McCambridge K, Wong H, Rauh MJ, Feilotter H, Quest G. Multiplex PCR for the Rapid Diagnosis of Myeloproliferative Neoplasms. Methods Mol Biol 2025. [PMID: 40227493 DOI: 10.1007/7651_2025_625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Myeloproliferative neoplasms (MPN) develop as a disease of aging, due to the disruption of normal hematopoiesis, triggered by somatic mutations and a sustained inflammatory cycle. Driving variants within JAK2, MPL, and CALR are associated with various subtypes of MPN, with known targeted therapeutics and guided patient prognosis. Targeted clinical testing for the diagnosis of these precise driving variants within these key genes has an integral role for personalized medicine.
Collapse
Affiliation(s)
- Laura Semenuk
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada.
- Department of Clinical Laboratory Services, Kingston Health Sciences Centre, Kingston, ON, Canada.
| | - Alissa Palazzolo
- Department of Clinical Laboratory Services, Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Kristen McCambridge
- Department of Clinical Laboratory Services, Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Henry Wong
- Department of Clinical Laboratory Services, Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Michael J Rauh
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Harriet Feilotter
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
- Department of Clinical Laboratory Services, Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Graeme Quest
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada.
- Department of Clinical Laboratory Services, Kingston Health Sciences Centre, Kingston, ON, Canada.
| |
Collapse
|
8
|
Pan Y, Chen L, Jiang Q, Chen D, Wu Y, Hou L, Lang H, Yan J. Research trends in essential thrombocythemia from 2001 to 2024: a bibliometric analysis. Discov Oncol 2025; 16:528. [PMID: 40232559 PMCID: PMC11999923 DOI: 10.1007/s12672-025-02232-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
OBJECTIVE This study aims to conduct a comprehensive bibliometric analysis of ET research, focusing on contributions from authors, institutions, and countries or regions, while mapping collaboration networks. Furthermore, it identifies development trends to provide insights for future research. METHODS A bibliometric analysis of ET-related publications (2001-2024) was conducted using data from the Web of Science Core Collection, focusing on publication trends, co-authorship networks, co-citation relationships, and citation bursts. RESULTS A total of 4,297 studies published in 778 journals were included in the analysis. ET research has grown rapidly, with major contributions from researchers in the United States and Europe, particularly through extensive collaborations. Leading figures such as Ayalew Tefferi and Alessandro M. Vannucchi have driven advances in ET classification, molecular mechanisms, and targeted therapies. The discovery of driver mutations, such as JAK2, has revolutionized the diagnostic and therapeutic approaches to ET. Research focus has shifted from clinical morphological diagnosis to molecular diagnostics, with the field now entering the era of targeted therapies. However, the heterogeneity of ET, the limitations of targeted therapies, particularly the lack of management experience and data for high-risk and special populations, as well as the incomplete understanding of the role of inflammation in the disease mechanism, continue to hinder both clinical and scientific progress in ET research. CONCLUSIONS Bibliometric analysis demonstrates significant advances in ET research, particularly in molecular pathology and targeted therapies. Future research should address ET heterogeneity, optimize management of high-risk and special populations, overcome the limitations of targeted therapies, and further elucidate the role of inflammation to achieve individualized precision therapy.
Collapse
Affiliation(s)
- Yiming Pan
- Department of Hematology, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Lingyan Chen
- School of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qun Jiang
- Department of Hematology, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dejian Chen
- School of Nursing, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanqin Wu
- Department of Hematology, Kunming Hospital of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Yunnan, China
| | - Li Hou
- Department of Hematology, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Haiyan Lang
- Department of Hematology, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China.
| | - Jun Yan
- Department of Respiratory Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
9
|
Laganà A, Ielo C, Buffolino S, Di Lascio A, Bizzoni L, Scalzulli E, Carmosino I, Bisegna ML, Diverio D, Breccia M. A novel complex mutation in the exon 9 of CALR gene: p.E381fs*52 (gagg_CCTCTTTGCCTC). Curr Res Transl Med 2025; 73:103514. [PMID: 40250195 DOI: 10.1016/j.retram.2025.103514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Affiliation(s)
- Alessandro Laganà
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Claudia Ielo
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Sonia Buffolino
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Attilio Di Lascio
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Luisa Bizzoni
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Emilia Scalzulli
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Ida Carmosino
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Maria Laura Bisegna
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Daniela Diverio
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Massimo Breccia
- Hematology, Department of Translational and Precision Medicine, Policlinico Umberto I-Sapienza University, Rome, Italy.
| |
Collapse
|
10
|
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
|
11
|
Hochman MJ, Vale CA, Hunter AM. SOHO State of the Art Updates and Next Questions | Choosing and Properly Using a JAK Inhibitor in Myelofibrosis. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:226-239. [PMID: 39358153 DOI: 10.1016/j.clml.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024]
Abstract
Myelofibrosis (MF) is a chronic myeloid neoplasm characterized by myeloproliferation, bone marrow fibrosis, splenomegaly, and constitutional symptoms related to pro-inflammatory cytokine signaling. Biologically, MF is characterized by constitutive activation of JAK-STAT signaling; accordingly, JAK inhibitors have been rationally developed to treat MF. Following the initial approval of ruxolitinib in 2011, three additional agents have been approved: fedratinib, pacritinib, and momelotinib. As these therapies are noncurative, allogeneic stem cell transplantation remains a key treatment modality and patients with MF who are deemed candidates should be referred to a transplant center. This potentially curative but toxic approach is typically reserved for patients with higher-risk disease, and JAK inhibitors are recommended in the pretransplant setting. JAK inhibitors have proven effective at managing splenomegaly and constitutional symptoms and should be started early in the disease course in patients presenting with these clinical manifestations; asymptomatic patients may initially be followed with close surveillance. Drug-related myelosuppression has been a challenge with initial JAK inhibitors, particularly in patients presenting with a cytopenic phenotype. However, newer agents, namely pacritinib and momelotinib, have proven more effective in this setting and are approved for patients with significant thrombocytopenia and anemia, respectively. Resistance or disease progression is clinically challenging and may be defined by several possible events, such as increasing splenomegaly or progression to accelerated or blast phase disease. However, with multiple JAK inhibitors now approved, sequencing of these agents appears poised to improve outcomes. Additionally, novel JAK inhibitors and JAK inhibitor-based combinations are in clinical development.
Collapse
Affiliation(s)
- Michael J Hochman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Colin A Vale
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Anthony M Hunter
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA.
| |
Collapse
|
12
|
Tarburn EL, Iversen L, Robertson C, McShane C, Duncombe A, McMullin MF, Harrison C, Mesa R, Anderson LA. Pathways to myeloproliferative neoplasm presentation and time to diagnosis: results from a cross-sectional study. BJGP Open 2025; 9:BJGPO.2024.0068. [PMID: 39079744 DOI: 10.3399/bjgpo.2024.0068] [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: 03/14/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Early cancer recognition is key to improving patient outcomes. Diagnosis is often delayed in patients with myeloproliferative neoplasms (MPNs), putting them at risk of thromboembolic events and other complications pre-diagnosis. A clear understanding of the barriers to presentation and diagnosis is required. AIM To explore barriers and factors influencing delayed presentation and diagnosis of MPNs. DESIGN & SETTING A cross-sectional study of patients with MPN within the UK and the Republic of Ireland. METHOD An online cross-sectional survey of patients with MPN was undertaken. Symptoms and factors influencing patient and GP delay were examined. Adjusted odds ratios (aORs) were calculated to explore the relationship between these factors and patient and GP delay. RESULTS Most (80.2%) of the 620 patients completing the survey reported symptomatic presentation. The most common symptoms associated with patient delay were pruritus (aOR 1.89, 95% confidence interval [CI] = 1.19 to 3.01), headaches (aOR 1.86, 95% CI = 1.13 to 2.82), and concentration difficulties (aOR 1.75, 95% CI = 1.12 to 2.76). Attributing symptoms to ageing (aOR 1.92, 95% CI = 1.19 to 3.11) and not wanting to burden the GP (2.04, 95% CI = 1.24 to 3.39) were significantly associated with patient delay. Those reporting >3 blood cancer warning signs were more likely to experience GP delay than those experiencing fewer (aOR 3.26, 95% CI = 1.75 to 6.29), and lack of relational continuity of GP care was significantly associated with GP delay (aOR 3.41, 95% CI = 1.65 to 7.28). CONCLUSION Debunking misconceptions around ageing, encouraging timely communication with GPs, and improving relational continuity of GP care could assist in reducing diagnostic delays, prevent potentially fatal disease complications, and ultimately improve outcomes for patients with MPN.
Collapse
Affiliation(s)
- Emma-Louise Tarburn
- Aberdeen Centre for Health Data Science, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Lisa Iversen
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | | | - Charlene McShane
- Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Andrew Duncombe
- Haematology Department, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Mary-Frances McMullin
- Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Claire Harrison
- Haematology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ruben Mesa
- Atrium Health Levine Cancer Institute, Charlotte, NC, US
| | - Lesley A Anderson
- Aberdeen Centre for Health Data Science, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
13
|
Mosnier C, Bellal S, Cottin L, Boyer F, Lemoine S, Bachelot A, Argentin J, Pawlicki B, Copin MC, Jouanneau-Courville R, Malinge A, Riou J, Hunault-Berger M, Ugo V, Orvain C, Luque Paz D. Relationship between additional mutations at diagnosis and treatment response in patients with essential thrombocythemia. Blood Adv 2025; 9:1303-1311. [PMID: 39820709 PMCID: PMC11950951 DOI: 10.1182/bloodadvances.2024014791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025] Open
Abstract
ABSTRACT Patients with essential thrombocythemia (ET) have a chronic evolution with a risk of hematologic transformation associated with a dismal outcome. Because patients with resistance or intolerance have adverse prognosis, it is important to identify which patient will respond to first-line treatment. We, therefore, aim to describe the association between additional mutations and response to first-line treatment in patients with ET. In this retrospective study, we analyzed the molecular landscape of 121 ET patients first-line treated with hydroxyurea (HU; n = 86) or pegylated interferon (peg-IFN; n = 35). Patients undergoing peg-IFN therapy were younger and had higher proportion of low and very low risk of thrombosis recurrence. A total of 62 patients (51%) had ≥1 additional mutations at diagnosis. At 12 months of treatment, 75 patients (62%) achieved complete response (CR), 37 (31%) partial response, and 7 (6%) no response. The presence of at least 1 additional mutation at diagnosis was associated with not achieving CR (hazard ratio [HR], 0.65; P = .038), whereas treatment with peg-IFN was associated with higher CR (HR, 2.00; P = .002). The number of additional mutations at diagnosis was associated with hematologic progressions (P < .0001). None of the patients receiving peg-IFN therapy progressed to myelofibrosis, whereas 16 of 86 patients (19%) treated with HU developed secondary myelofibrosis. In conclusion, our results suggest that the presence of at least 1 additional mutation at diagnosis is associated with failure to achieve CR and also with an increased risk of hematologic evolution.
Collapse
Affiliation(s)
- Carole Mosnier
- Univ Angers, Nantes Université, CHU Angers, INSERM, CNRS, CRCI2NA, Angers, France
- Maladies du Sang, CHU d'Angers, Angers, France
| | - Sarah Bellal
- Service de Pathologie, Univ Angers, CHU Angers, Angers, France
| | - Laurane Cottin
- Univ Angers, Nantes Université, CHU Angers, INSERM, CNRS, CRCI2NA, Angers, France
- Laboratoire d'Hématologie, CHU Angers, Angers, France
| | | | - Sandrine Lemoine
- Univ Angers, Nantes Université, CHU Angers, INSERM, CNRS, CRCI2NA, Angers, France
| | | | | | | | - Marie-Christine Copin
- Univ Angers, Nantes Université, CHU Angers, INSERM, CNRS, CRCI2NA, Angers, France
- Service de Pathologie, Univ Angers, CHU Angers, Angers, France
| | | | - Anaïs Malinge
- Laboratoire d'Hématologie, CHU Angers, Angers, France
| | - Jérémie Riou
- Methodology and Biostatistics Department, Delegation to Clinical Research and Innovation, Angers University Hospital, Angers, France
| | - Mathilde Hunault-Berger
- Univ Angers, Nantes Université, CHU Angers, INSERM, CNRS, CRCI2NA, Angers, France
- Maladies du Sang, CHU d'Angers, Angers, France
- Fédération Hospitalo-Universitaire Grand-Ouest Against Leukemia (GOAL), Angers, France
| | - Valérie Ugo
- Univ Angers, Nantes Université, CHU Angers, INSERM, CNRS, CRCI2NA, Angers, France
- Laboratoire d'Hématologie, CHU Angers, Angers, France
- Fédération Hospitalo-Universitaire Grand-Ouest Against Leukemia (GOAL), Angers, France
| | - Corentin Orvain
- Univ Angers, Nantes Université, CHU Angers, INSERM, CNRS, CRCI2NA, Angers, France
- Maladies du Sang, CHU d'Angers, Angers, France
- Fédération Hospitalo-Universitaire Grand-Ouest Against Leukemia (GOAL), Angers, France
| | - Damien Luque Paz
- Univ Angers, Nantes Université, CHU Angers, INSERM, CNRS, CRCI2NA, Angers, France
- Laboratoire d'Hématologie, CHU Angers, Angers, France
- Fédération Hospitalo-Universitaire Grand-Ouest Against Leukemia (GOAL), Angers, France
| |
Collapse
|
14
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
15
|
Resar LMS, Luo LZ. High Mobility Group A1 Chromatin Keys: Unlocking the Genome During MPN Progression. Int J Mol Sci 2025; 26:2125. [PMID: 40076747 PMCID: PMC11899949 DOI: 10.3390/ijms26052125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/11/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Patients with chronic, indolent myeloproliferative neoplasms (MPNs) are at risk for transformation to highly lethal leukemia, although targetable mechanisms driving progression remain elusive. We discovered that the High Mobility Group A1 (HMGA1) gene is up-regulated with MPN progression in patients and required for evolution into myelofibrosis (MF) or acute myeloid leukemia (AML) in preclinical models. HMGA1 encodes the HMGA1 epigenetic regulators that modulate the chromatin state during embryogenesis and tissue regeneration. While HMGA1 is silenced in most differentiated cells, it becomes aberrantly re-expressed in JAK2 mutant (JAK2-V617F) MPN, with the highest levels after transformation to secondary MF or AML. Here, we review recent work highlighting HMGA1 function in MPN progression. Though underlying mechanisms continue to emerge, increasing evidence suggests that HMGA1 functions as a "chromatin key" required to "unlock" regions of the genome involved in clonal expansion and progression in MPN. Together, these findings illuminate HMGA1 as a driver of MPN progression and a promising therapeutic target.
Collapse
Affiliation(s)
- Linda M. S. Resar
- Departments of Medicine (Hematology), Oncology, Pathology and Institute for Cellular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | | |
Collapse
|
16
|
Williams MJ, Wang X, Bastos HP, Grondys-Kotarba G, Wu Q, Jin S, Johnson C, Mende N, Calderbank E, Wantoch M, Park HJ, Mantica G, Hannah R, Wilson NK, Pask DC, Hamilton TL, Kinston SJ, Asby R, Sneade R, Baxter EJ, Campbell P, Vassiliou GS, Laurenti E, Li J, Göttgens B, Green AR. Maintenance of hematopoietic stem cells by tyrosine-unphosphorylated STAT5 and JAK inhibition. Blood Adv 2025; 9:291-309. [PMID: 39374575 PMCID: PMC7617191 DOI: 10.1182/bloodadvances.2024014046] [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: 06/20/2024] [Revised: 09/04/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
ABSTRACT Adult hematopoietic stem cells (HSCs) are responsible for the lifelong production of blood and immune cells, a process regulated by extracellular cues, including cytokines. Many cytokines signal through the conserved Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway in which tyrosine-phosphorylated STATs (pSTATs) function as transcription factors. STAT5 is a pivotal downstream mediator of several cytokines known to regulate hematopoiesis, but its function in the HSC compartment remains poorly understood. In this study, we show that STAT5-deficient HSCs exhibit an unusual phenotype, including reduced multilineage repopulation and self-renewal, combined with reduced exit from quiescence and increased differentiation. This was driven not only by the loss of canonical pSTAT5 signaling, but also by the loss of distinct transcriptional functions mediated by STAT5 that lack canonical tyrosine phosphorylation (uSTAT5). Consistent with this concept, expression of an unphosphorylatable STAT5 mutant constrained wild-type HSC differentiation, promoted their maintenance, and upregulated transcriptional programs associated with quiescence and stemness. The JAK1/2 inhibitor, ruxolitinib, which increased the uSTAT5:pSTAT5 ratio, had similar effects on murine HSC function; it constrained HSC differentiation and proliferation, promoted HSC maintenance, and upregulated transcriptional programs associated with stemness. Ruxolitinib also enhanced serial replating of normal human hematopoietic stem and progenitor cells (HSPCs), calreticulin-mutant murine HSCs, and HSPCs obtained from patients with myelofibrosis. Our results therefore reveal a previously unrecognized interplay between pSTAT5 and uSTAT5 in the control of HSC function and highlight JAK inhibition as a potential strategy for enhancing HSC function during ex vivo culture. Increased levels of uSTAT5 may also contribute to the failure of JAK inhibitors to eradicate myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Matthew J. Williams
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Xiaonan Wang
- Department of Public Health, School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hugo P. Bastos
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | | | - Qin Wu
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Shucheng Jin
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Carys Johnson
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Nicole Mende
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Emily Calderbank
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Michelle Wantoch
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Hyun Jung Park
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Giovanna Mantica
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Rebecca Hannah
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Nicola K. Wilson
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Dean C. Pask
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Tina L. Hamilton
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Sarah J. Kinston
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Ryan Asby
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Rachel Sneade
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - E. Joanna Baxter
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Peter Campbell
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Cancer Genomics, Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - George S. Vassiliou
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Elisa Laurenti
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Juan Li
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Berthold Göttgens
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Anthony R. Green
- Department of Haematology, Wellcome–Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
17
|
Schäfersküpper M, Simon A, Yorgan TA, von Brackel FN, Delsmann MM, Baranowsky A, Gagelmann N, Ayuk F, Schinke T, Amling M, Kröger N, Rolvien T. Reconstructing skeletal homeostasis through allogeneic hematopoietic stem cell transplantation in myelofibrosis. Nat Commun 2025; 16:741. [PMID: 39833182 PMCID: PMC11747566 DOI: 10.1038/s41467-025-55915-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
Myeloproliferative neoplasm-associated myelofibrosis is a clonal stem cell process characterized by pronounced bone marrow fibrosis associated with extramedullary hematopoiesis and splenomegaly. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) represents the only curative treatment leading to bone marrow fibrosis regression. Here we provide an in-depth skeletal characterization of myelofibrosis patients before and after allo-HSCT utilizing clinical high-resolution imaging, laboratory analyses, and bone biopsy studies. Despite unimpaired bone microarchitecture at peripheral skeletal sites, we observe a marked increase in bone mineral density at the lumbar spine and proximal femur, which is histologically related to severe bone marrow fibrosis and osteosclerosis, fully normalizing after allo-HSCT. Importantly, the regression of fibrosis is accompanied by vanishing osteosclerosis along with restored osteoclastic resorption activity and whole-body calcium homeostasis. Together, our results provide evidence for an extensive reconstruction of skeletal homeostasis by allo-HSCT in MF, leading to rapid resolution of osteosclerosis.
Collapse
Affiliation(s)
- Mathias Schäfersküpper
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Simon
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timur A Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix N von Brackel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian M Delsmann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Baranowsky
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nico Gagelmann
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
18
|
Okura GC, Bharadwaj AG, Waisman DM. Calreticulin-From the Endoplasmic Reticulum to the Plasma Membrane-Adventures of a Wandering Protein. Cancers (Basel) 2025; 17:288. [PMID: 39858072 PMCID: PMC11764459 DOI: 10.3390/cancers17020288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Calreticulin (CRT) is a 46 kDa highly conserved protein initially identified as calregulin, a prominent Ca2+-binding protein of the endoplasmic reticulum (ER). Subsequent studies have established that CRT functions in the ER's protein folding response and Ca2+ homeostatic mechanisms. An ER retention signal on the carboxyl terminus of CRT suggested that CRT was restricted to the ER. However, the identification of CRT in the nucleus and cytosol has established that CRT is a multi-compartmental, multifunctional protein. CRT also plays an important role in cancer progression. Most recently, CRT was identified on the cell surface and shown to be a potent 'eat-me' signal that plays a key role in the uptake of apoptotic and viable cancer cells by phagocytes. Elevated CRT exposure on the outer leaflet of cancer cells has been linked with anticancer immunity and superior therapeutic outcomes in patients with non-small cell lung carcinoma, colorectal carcinoma, acute myeloid leukemia, ovarian cancer, and high-grade serous carcinomas. Mutations in the CRT gene have been identified in a subset of patients with myeloproliferative neoplasms. The most recent studies from our laboratory have revealed a new and significant function for extracellular CRT as a plasminogen receptor. This discovery has profound implications for our understanding of the role of CRT in myeloproliferative neoplasms, specifically, essential thrombocythemia.
Collapse
Affiliation(s)
- Gillian C. Okura
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (G.C.O.); (A.G.B.)
| | - Alamelu G. Bharadwaj
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (G.C.O.); (A.G.B.)
| | - David M. Waisman
- Department of Pathology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (G.C.O.); (A.G.B.)
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 1X5, Canada
| |
Collapse
|
19
|
Hu X, Yu X, Zhang L, Zhang Q, Ji M, Qi K, Wang S, Li Z, Xu K, Fu C. The aberrantly activated AURKB supports and complements the function of AURKA in CALR mutated cells through regulating the cell growth and differentiation. Exp Cell Res 2025; 444:114377. [PMID: 39706286 DOI: 10.1016/j.yexcr.2024.114377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/12/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Aurora kinase B (AURKB) was reported to assist Aurora kinase A (AURKA) to regulate cellular mitosis. AURKA has been found activated in myeloproliferative neoplasms (MPNs) patients with CALR gene mutation, however, it's unclear whether AURKB displays a compensatory function of AURKA in regulation of CALR mutant cell growth and differentiation. Here, we found that AURKB, similar with AURKA, was aberrantly activated in CALR mutant patients, and displayed a more tolerance to the aurora kinase inhibitor. Inhibition of AURKA decreased cell growth and colony formation, induced cell differentiation and apoptosis, while, this inhibitive degree was further enhanced when AURKB was blocked by incremental inhibitor. Transcriptomic analyses revealed a more significant gene enrichment in cells with knockdown of AURKB than that of AURKA, mainly reflecting in oxidative phosphorylation, mitosis, proliferation and apoptosis signaling pathway. Moreover, downregulation of AURKB enhanced cell growth arrest and apoptosis more obviously than that of AURKA, and additionally promoted cell differentiation and metabolism-oxygen consumption rate (OCR). Otherwise, overexpression of AURKA or AURKB facilitated the cell proliferation of CALR mutant cells, and made cells more sensitive to the aurora kinase inhibitor. These results suggest that activated AURKB not only supports the functions of AURKA in promoting the growth of CALR mutated cells, but also has impeded the differentiation of these cells.
Collapse
Affiliation(s)
- Xueting Hu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Xiangru Yu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Liwei Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Qigang Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Mengchu Ji
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kunming Qi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Shujin Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.
| | - Chunling Fu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
20
|
De Moner B, Martinez-Sanchez J, Garrote M, Ramos A, Ventosa-Capell H, Moreno-Castaño A, Nomdedeu M, Ojeda A, Escolar G, Garcia-Pagan JC, Arellano-Rodrigo E, Carreras E, Alvarez-Larran A, Díaz-Ricart M. Endothelial Damage in JAK2V617F Myeloproliferative Neoplasms with Splanchnic Vein Thrombosis. Thromb Haemost 2025. [PMID: 39809451 DOI: 10.1055/a-2498-4849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
BACKGROUND JAK2V617F-mutated myeloproliferative neoplasms (MPN) exhibit abnormal proliferation of bone marrow progenitors and increased risk of thrombosis, specifically in splanchnic veins (SVT). The contribution of the endothelium to the development of the prothrombotic phenotype was explored. MATERIAL AND METHODS Plasma and serum samples from JAK2V617F MPN patients with (n=26) or without (n=7) thrombotic debut and different treatments, were obtained (n=33). Cultured endothelial cells (ECs) were exposed to serum samples from these patients and from healthy donors as controls. Changes in markers of inflammation (VCAM-1, ICAM-1), cell permeability (VE-cadherin), production of VWF, extracellular matrix (ECM) reactivity, and activation of intracellular signaling pathways related to stress, proliferation, inflammation (Akt, p44/42, IkBa), and JAK2/STAT3 pathway, were assessed by immunofluorescence, flow adhesion, SDS-PAGE and immunoblot. Additionally, circulating markers of endothelial activation and damage (VWF, sVCAM-1, sTNFRI, thrombomodulin, angiopoietin-2, a2-antiplasmin activity, PAI-1) were evaluated in Patients' plasma. RESULTS The in vitro studies showed that EC exposure to MPN thrombotic patients' sera resulted in increased VCAM-1 and ICAM-1, and reduced VE-cadherin expression (p<0.05) at the cell surface. Production and release of VWF to the ECM were higher (p<0.05), with increased platelet adhesion after perfusing whole blood, being more noticeable in response to sera from non-treated patients. Furthermore, intracellular activation of Akt, p44/42, IkBa and JAK2/STAT3 was observed. Moreover, plasma levels of VWF, TNF-R1, VCAM-1, thrombomodulin, and angiopoietin-2 were higher in JAK2V617F+ MPN patients with thrombosis. CONCLUSION The present findings suggest that circulating factors in MPNs with SVT debut induce endothelial proinflammatory and prothrombotic phenotypes, which are modulated in vitro with MPN treatment.
Collapse
Affiliation(s)
- Blanca De Moner
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, University of Barcelona, Hospital Clínic de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Julia Martinez-Sanchez
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Marta Garrote
- Hematopathology Section, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, IDIBAPS, Spain
| | - Alex Ramos
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | | | - Ana Moreno-Castaño
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Meritxell Nomdedeu
- Hemostasis and Hemotherapy Department, Institut del Càncer i Malalties de la Sang (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Asunción Ojeda
- Liver Unit, Barcelona Hepatic Haemodynamic Laboratory, Hospital Clínic de Barcelona, IDIBAPS and CIBERehd, Barcelona, Spain
| | - Gines Escolar
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Joan Carles Garcia-Pagan
- Liver Unit, Barcelona Hepatic Haemodynamic Laboratory, Hospital Clínic de Barcelona, IDIBAPS and CIBERehd, Barcelona, Spain
| | - Eduardo Arellano-Rodrigo
- Hemostasis and Hemotherapy Department, Institut del Càncer i Malalties de la Sang (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Enric Carreras
- Barcelona Endothelium Team, Barcelona, Spain
- Fundació Carreras contra la Leucèmia, Barcelona, Spain
| | - Alberto Alvarez-Larran
- Hematology Department, Institut del Càncer i Malalties de la Sang (ICAMS), Hospital Clínic de Barcelona, IDIBAPS, Barcelona, Spain
| | - Maribel Díaz-Ricart
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| |
Collapse
|
21
|
Faiz M, Riedemann M, Jutzi JS, Mullally A. Mutant Calreticulin in MPN: Mechanistic Insights and Therapeutic Implications. Curr Hematol Malig Rep 2025; 20:4. [PMID: 39775969 DOI: 10.1007/s11899-024-00749-4] [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] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW More than a decade following the discovery of Calreticulin (CALR) mutations as drivers of myeloproliferative neoplasms (MPN), advances in the understanding of CALR-mutant MPN continue to emerge. Here, we summarize recent advances in mehanistic understanding and in targeted therapies for CALR-mutant MPN. RECENT FINDINGS Structural insights revealed that the mutant CALR-MPL complex is a tetramer and the mutant CALR C-terminus is exposed on the cell surface. Targeting mutant CALR utilizing antibodies is the leading therapeutic approach, while mutant CALR-directed vaccines are also in early clinical trials. Additionally, chimeric antigen receptor (CAR) T-cells directed against mutant CALR are under evaluation in preclinical models. Approaches addressing the cellular effects of mutant CALR beyond MPL-JAK-STAT activation, such as targeting the unfolded protein response, proteasome, and N-glycosylation pathways, have been tested in preclinical models. In CALR-mutant MPN, the path from discovery to mechanistic understanding to direct therapeutic targeting has advanced rapidly. The longer-term goal remains clonally-selective therapies that modify the disease course in patients.
Collapse
Affiliation(s)
- Mifra Faiz
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Institute of Medicine, Boston, MA, 02115, USA
| | - Merle Riedemann
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Institute of Medicine, Boston, MA, 02115, USA
| | - Jonas S Jutzi
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Institute of Medicine, Boston, MA, 02115, USA
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Hematology Division, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA.
| |
Collapse
|
22
|
Yamamoto Y, Iba S, Inaguma Y, Okamoto A, Abe A. ELC52: a novel megakaryocytic leukemia cell line with a CALR type 1 mutation. Leukemia 2025; 39:234-237. [PMID: 39379530 DOI: 10.1038/s41375-024-02434-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Affiliation(s)
- Yukiya Yamamoto
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Aichi, Japan.
- School of Medicine, Fujita Health University, Toyoake, Aichi, Japan.
| | - Sachiko Iba
- Department of Hematology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoko Inaguma
- Department of Clinical General Medicine, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Akinao Okamoto
- Department of Hematology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Akihiro Abe
- Department of Hematology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
23
|
Hoermann G, Khoury JD. Can molecular patterns help to classify overlapping entities in myeloid neoplasms? Histopathology 2025; 86:146-157. [PMID: 39428913 PMCID: PMC11648353 DOI: 10.1111/his.15339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Myeloid neoplasms include myeloproliferative and myelodysplastic neoplasms and acute myeloid leukaemia. Historically, these diseases have been diagnosed based on clinicopathological features with sometimes arbitrary thresholds that have persisted even as molecular features were gradually incorporated into their classification. As such, although current diagnostic approaches can classify the majority of myeloid neoplasms accurately using a combination of molecular and clinicopathological features, some areas of overlap persist and occasionally pose diagnostic challenges. These include overlap across BCR::ABL1-negative myeloproliferative neoplasms; between clonal cytopenia of undetermined significance and myelodysplastic neoplasms; myelodysplastic/myeloproliferative neoplasms; and, detection of KIT mutations in myeloid neoplasms other than mastocytosis, raising the prospect of systemic mastocytosis. Molecular testing has become state of the art in the diagnostic work-up of myeloid neoplasms, and molecular patterns can inherently help to classify overlapping entities if considered within a framework of haematological presentations. For future development, molecular testing will likely include whole genome and transcriptome sequencing, and primarily molecular classifications of myeloid neoplasms have already been suggested. As such, genetically defined groups should still constitute the basis for our understanding of disease development from early onset to progression, while clinicopathological features could then be used to describe the stage of the disease rather than the specific type of myeloid neoplasm.
Collapse
Affiliation(s)
| | - Joseph D Khoury
- Department of Pathology, Microbiology, and ImmunologyUniversity of Nebraska Medical CenterOmahaUSA
| |
Collapse
|
24
|
Ozygała A, Rokosz-Mierzwa J, Widz P, Skowera P, Wiliński M, Styka B, Lejman M. Biological Markers of Myeloproliferative Neoplasms in Children, Adolescents and Young Adults. Cancers (Basel) 2024; 16:4114. [PMID: 39682300 DOI: 10.3390/cancers16234114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs) are clonal hematopoietic cancers characterized by hyperproliferation of the myeloid lineages. These clonal marrow disorders are extremely rare in pediatric patients. MPN is reported to occur 100 times more frequently in adults, and thus research is primarily focused on this patient group. At present, modern diagnostic techniques, primarily genetic, facilitate the identification of the biology of these diseases. The key genes are JAK2, MPL, and CALR, namely, driver mutations, which are present in approximately 90% of patients with suspected MPN. Moreover, there are more than 20 other mutations that affect the development of these hematological malignancies, as evidenced by a review of the literature. The pathogenic mechanism of MPNs is characterized by the dysregulation of the JAK/STAT signaling pathway (JAK2, MPL, CALR), DNA methylation (TET2, DNMT3A, IDH1/2), chromatin structure (ASXL1, EZH2), and splicing (SF3B1, U2AF2, SRSF2). Although rare, myeloproliferative neoplasms can involve young patients and pose unique challenges for clinicians in diagnosis and therapy. The paper aims to review the biological markers of MPNs in pediatric populations-a particular group of patients that has been poorly studied due to the low frequency of MPN diagnosis.
Collapse
Affiliation(s)
- Aleksandra Ozygała
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Joanna Rokosz-Mierzwa
- Department of Genetic Diagnostics, University Children's Hospital, 20-093 Lublin, Poland
| | - Paulina Widz
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-059 Lublin, Poland
| | - Paulina Skowera
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Mateusz Wiliński
- Department of Genetic Diagnostics, University Children's Hospital, 20-093 Lublin, Poland
| | - Borys Styka
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
25
|
Puli'uvea C, Immanuel T, Green TN, Tsai P, Shepherd PR, Kalev-Zylinska ML. Insights into the role of JAK2-I724T variant in myeloproliferative neoplasms from a unique cohort of New Zealand patients. Hematology 2024; 29:2297597. [PMID: 38197452 DOI: 10.1080/16078454.2023.2297597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
OBJECTIVES This study aimed to compile bioinformatic and experimental information for JAK2 missense variants previously reported in myeloproliferative neoplasms (MPN) and determine if germline JAK2-I724T, recently found to be common in New Zealand Polynesians, associates with MPN. METHODS For all JAK2 variants found in the literature, gnomAD_exome allele frequencies were extracted and REVEL scores were calculated using the dbNSFP database. We investigated the prevalence of JAK2-I724T in a cohort of 111 New Zealand MPN patients using a TaqMan assay, examined its allelic co-occurrence with JAK2-V617F using Oxford Nanopore sequencing, and modelled the impact of I724T on JAK2 using I-Mutant and ChimeraX software. RESULTS Several non-V617F JAK2 variants previously reported in MPN had REVEL scores greater than 0.5, suggesting pathogenicity. JAK2-I724T (REVEL score 0.753) was more common in New Zealand Polynesian MPN patients (n = 2/27; 7.4%) than in other New Zealand patients (n = 0/84; 0%) but less common than expected for healthy Polynesians (n = 56/377; 14.9%). Patients carrying I724T (n = 2), one with polycythaemia vera and one with essential thrombocythaemia, had high-risk MPN. Both patients with JAK2-I724T were also positive for JAK2-V617F, found on the same allele as I724T, as well as separately. In silico modelling did not identify noticeable structural changes that would give JAK2-I724T a gain-of-function. CONCLUSION Several non-canonical JAK2 variants with high REVEL scores have been reported in MPN, highlighting the need to further understand their relationship with disease. The JAK2-I724T variant does not drive MPN, but additional investigations are required to exclude any potential modulatory effect on the MPN phenotype.
Collapse
Affiliation(s)
- Christopher Puli'uvea
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Hosted by the University of Auckland, Auckland, New Zealand
| | - Tracey Immanuel
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Taryn N Green
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Peter Tsai
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Hosted by the University of Auckland, Auckland, New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Hosted by the University of Auckland, Auckland, New Zealand
| | - Maggie L Kalev-Zylinska
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Pathology and Laboratory Medicine, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
26
|
Thaw K, Harrison CN, Sriskandarajah P. JAK Inhibitors for Myelofibrosis: Strengths and Limitations. Curr Hematol Malig Rep 2024; 19:264-275. [PMID: 39400853 PMCID: PMC11567979 DOI: 10.1007/s11899-024-00744-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE OF REVIEW The landscape of myelofibrosis (MF) has changed since the discovery of the JAK2 V617F mutation and subsequent development of JAK inhibitors (JAKis). However, treatment with JAKis remain a challenge. In this review we critically analyze the strengths and limitations of currently available JAK inhibitors. RECENT FINDINGS In MF patients, JAK inhibitors have been associated with reduced symptom burden and spleen size, as well as improved survival. However, durability of response and development of treatment resistance remain an issue. Recently, there has been increased efforts to optimize treatment with the development of highly selective JAK inhibitors, as well as use of combination agents to counter disease resistance through targeting aberrant signaling pathways. Treatment of MF patients with JAKi therapy can be challenging but the development of more potent and selective JAK inhibitors, as well as combination therapies, represent exciting treatment advances in this field.
Collapse
Affiliation(s)
- K Thaw
- Department of Haematology, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - C N Harrison
- Department of Haematology, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.
| | - P Sriskandarajah
- Department of Haematology, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| |
Collapse
|
27
|
Benlabiod C, Psaila B. INCA033989: the first shot on goal for MPNs? Blood 2024; 144:2278-2279. [PMID: 39607716 DOI: 10.1182/blood.2024026811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
ABSTRACT In this issue of Blood, Reis et al1 identify a monoclonal antibody, INCA033989, that selectively targets mutant calreticulin (mutCALR) in myeloproliferative neoplasms (MPNs), inhibiting its oncogenic activity without affecting normal hematopoiesis.
Collapse
Affiliation(s)
| | - Bethan Psaila
- University of Oxford
- Oxford University Hospitals NHS Trust
| |
Collapse
|
28
|
Kędzierska M, Bańkosz M. Role of Proteins in Oncology: Advances in Cancer Diagnosis, Prognosis, and Targeted Therapy-A Narrative Review. J Clin Med 2024; 13:7131. [PMID: 39685591 DOI: 10.3390/jcm13237131] [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: 10/27/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Modern oncology increasingly relies on the role of proteins as key components in cancer diagnosis, prognosis, and targeted therapy. This review examines advancements in protein biomarkers across several cancer types, including breast cancer, lung cancer, ovarian cancer, and hepatocellular carcinoma. These biomarkers have proven critical for early detection, treatment response monitoring, and tailoring personalized therapeutic strategies. The article highlights the utility of targeted therapies, such as tyrosine kinase inhibitors and monoclonal antibodies, in improving treatment efficacy while minimizing systemic toxicity. Despite these advancements, challenges like tumor resistance, variability in protein expression, and diagnostic heterogeneity persist, complicating universal application. The review underscores future directions, including the integration of artificial intelligence, advanced protein analysis technologies, and the development of combination therapies to overcome these barriers and refine personalized cancer treatment.
Collapse
Affiliation(s)
- Magdalena Kędzierska
- Department of Chemotherapy, Medical University of Lodz, Copernicus Memorial Hospital of Lodz, 90-549 Lodz, Poland
| | - Magdalena Bańkosz
- CUT Doctoral School, Faculty of Materials Engineering and Physics, Department of Material Engineering, Cracow University of Technology, 37 Jana Pawla II Av., 31-864 Krakow, Poland
| |
Collapse
|
29
|
Dunn WG, McLoughlin MA, Vassiliou GS. Clonal hematopoiesis and hematological malignancy. J Clin Invest 2024; 134:e180065. [PMID: 39352393 PMCID: PMC11444162 DOI: 10.1172/jci180065] [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] [Indexed: 10/03/2024] Open
Abstract
Clonal hematopoiesis (CH), the expansion of hematopoietic stem cells and their progeny driven by somatic mutations in leukemia-associated genes, is a common phenomenon that rises in prevalence with advancing age to affect most people older than 70 years. CH remains subclinical in most carriers, but, in a minority, it progresses to a myeloid neoplasm, such as acute myeloid leukemia, myelodysplastic syndrome, or myeloproliferative neoplasm. Over the last decade, advances in our understanding of CH, its molecular landscape, and the risks associated with different driver gene mutations have culminated in recent developments that allow for a more precise estimation of myeloid neoplasia risk in CH carriers. In turn, this is leading to the development of translational and clinical programs to intercept and prevent CH from developing into myeloid neoplasia. Here, we give an overview of the spectrum of CH driver mutations, what is known about their pathophysiology, and how this informs the risk of incident myeloid malignancy.
Collapse
Affiliation(s)
- William G. Dunn
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Trust, Cambridge, United Kingdom
| | - Matthew A. McLoughlin
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - George S. Vassiliou
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Trust, Cambridge, United Kingdom
| |
Collapse
|
30
|
Carretta C, Parenti S, Bertesi M, Rontauroli S, Badii F, Tavernari L, Genovese E, Malerba M, Papa E, Sperduti S, Enzo E, Mirabile M, Pedrazzi F, Neroni A, Tombari C, Mora B, Maffioli M, Mondini M, Brociner M, Maccaferri M, Tenedini E, Martinelli S, Bartalucci N, Bianchi E, Casarini L, Potenza L, Luppi M, Tagliafico E, Guglielmelli P, Simoni M, Passamonti F, Norfo R, Vannucchi AM, Manfredini R. Chromosome 9p trisomy increases stem cells clonogenic potential and fosters T-cell exhaustion in JAK2-mutant myeloproliferative neoplasms. Leukemia 2024; 38:2171-2182. [PMID: 39179669 PMCID: PMC11436358 DOI: 10.1038/s41375-024-02373-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024]
Abstract
JAK2V617F is the most recurrent genetic mutation in Philadelphia-negative chronic Myeloproliferative Neoplasms (MPNs). Since the JAK2 locus is located on Chromosome 9, we hypothesized that Chromosome 9 copy number abnormalities may be a disease modifier in JAK2V617F-mutant MPN patients. In this study, we identified a subset of MPN patients with partial or complete Chromosome 9 trisomy (+9p patients), who differ from JAK2V617F-homozygous MPN patients as they carry three JAK2 alleles as well as three copies of all neighboring gene loci, including CD274, encoding immunosuppressive Programmed death-ligand 1 (PD-L1) protein. Investigation of the clonal hierarchy revealed that the JAK2V617F occurs first, followed by +9p. Functionally, CD34+ cells from +9p MPN patients demonstrated increased clonogenicity, generating a greater number of primitive colonies, due to high OCT4 and NANOG expression, with knock-down of these genes leading to a genotype-specific decrease in colony numbers. Moreover, our analysis revealed increased PD-L1 surface expression in malignant monocytes from +9p patients, while analysis of the T cell compartment unveiled elevated levels of exhausted cytotoxic T cells. Overall, here we identify a distinct novel subgroup of MPN patients, who feature a synergistic interplay between +9p and JAK2V617F that shapes immune escape characteristics and increased stemness in CD34+ cells.
Collapse
Affiliation(s)
- Chiara Carretta
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sandra Parenti
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Bertesi
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sebastiano Rontauroli
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Filippo Badii
- Department of Cancer Biology, Thomas Jefferson University and Sidney Kimmel Cancer Center, Philadelphia, PA, USA
| | - Lara Tavernari
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Genovese
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marica Malerba
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Papa
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Samantha Sperduti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Enzo
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Margherita Mirabile
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Pedrazzi
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anita Neroni
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Camilla Tombari
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Margherita Maffioli
- S.C. Ematologia, Ospedale di Circolo e Fondazione Macchi-ASST Sette Laghi, Varese, Italy
| | - Marco Mondini
- S.C. Ematologia, Ospedale di Circolo e Fondazione Macchi-ASST Sette Laghi, Varese, Italy
| | - Marco Brociner
- S.C. Ematologia, Ospedale di Circolo e Fondazione Macchi-ASST Sette Laghi, Varese, Italy
| | | | - Elena Tenedini
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Martinelli
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Niccolò Bartalucci
- CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence, AOU Careggi, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elisa Bianchi
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Leonardo Potenza
- Hematology Unit, Modena University Hospital, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Mario Luppi
- Hematology Unit, Modena University Hospital, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Paola Guglielmelli
- CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence, AOU Careggi, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Ruggiero Norfo
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Maria Vannucchi
- CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, University of Florence, AOU Careggi, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Rossella Manfredini
- Interdepartmental Centre for Stem Cells and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy.
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
31
|
Grasshoff M, Kalmer M, Chatain N, Kricheldorf K, Maurer A, Weiskirchen R, Koschmieder S, Costa IG. SIngle cell level Genotyping Using scRna Data (SIGURD). Brief Bioinform 2024; 25:bbae604. [PMID: 39559832 PMCID: PMC11574290 DOI: 10.1093/bib/bbae604] [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: 07/16/2024] [Revised: 10/14/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024] Open
Abstract
MOTIVATION By accounting for variants within measured transcripts, it is possible to evaluate the status of somatic variants using single-cell RNA-sequencing (scRNA-seq) and to characterize their clonality. However, the sparsity (very few reads per transcript) or bias in protocols (favoring 3' ends of the transcripts) makes the chance of capturing somatic variants very unlikely. This can be overcome by targeted sequencing or the use of mitochondrial variants as natural barcodes for clone identification. Currently, available computational tools focus on genotyping, but do not provide functionality for combined analysis of somatic and mitochondrial variants and functional analysis such as characterization of gene expression changes in detected clones. RESULTS Here, we propose SIGURD (SIngle cell level Genotyping Using scRna Data) (SIGURD), which is an R-based pipeline for the clonal analysis of scRNA-seq data. This allows the quantification of clones by leveraging both somatic and mitochondrial variants. SIGURD also allows for functional analysis after clonal detection: association of clones with cell populations, detection of differentially expressed genes across clones, and association of somatic and mitochondrial variants. Here, we demonstrate the power of SIGURD by analyzing single-cell data of colony-forming cells derived from patients with myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Martin Grasshoff
- Institute for Computational Genomics, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, NRW, Germany
| | - Milena Kalmer
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Pauwelstr 30, 52074 Aachen, NRW, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Pauwelstr 30, 52074 Aachen, NRW, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Kim Kricheldorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Pauwelstr 30, 52074 Aachen, NRW, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Angela Maurer
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Pauwelstr 30, 52074 Aachen, NRW, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University, Pauwelstr 30, 52074 Aachen, NRW, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Pauwelstr 30, 52074 Aachen, NRW, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, NRW, Germany
| |
Collapse
|
32
|
Schmidt D, Endres C, Hoefflin R, Andrieux G, Zwick M, Karantzelis N, Staehle HF, Vinnakota JM, Duquesne S, Mozaffari M, Pfeifer D, Becker H, Blazar BR, Zähringer A, Duyster J, Brummer T, Boerries M, Baumeister J, Shoumariyeh K, Li J, Green AR, Heidel FH, Tirosh I, Pahl HL, Leimkühler N, Köhler N, de Toledo MAS, Koschmieder S, Zeiser R. Oncogenic Calreticulin Induces Immune Escape by Stimulating TGFβ Expression and Regulatory T-cell Expansion in the Bone Marrow Microenvironment. Cancer Res 2024; 84:2985-3003. [PMID: 38885318 PMCID: PMC11405138 DOI: 10.1158/0008-5472.can-23-3553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 04/12/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
Increasing evidence supports the interplay between oncogenic mutations and immune escape mechanisms. Strategies to counteract the immune escape mediated by oncogenic signaling could provide improved therapeutic options for patients with various malignancies. As mutant calreticulin (CALR) is a common driver of myeloproliferative neoplasms (MPN), we analyzed the impact of oncogenic CALRdel52 on the bone marrow (BM) microenvironment in MPN. Single-cell RNA sequencing revealed that CALRdel52 led to the expansion of TGFβ1-producing erythroid progenitor cells and promoted the expansion of FoxP3+ regulatory T cells (Treg) in a murine MPN model. Treatment with an anti-TGFβ antibody improved mouse survival and increased the glycolytic activity in CD4+ and CD8+ T cells in vivo, whereas T-cell depletion abrogated the protective effects conferred by neutralizing TGFβ. TGFβ1 reduced perforin and TNFα production by T cells in vitro. TGFβ1 production by CALRdel52 cells was dependent on JAK1/2, PI3K, and ERK activity, which activated the transcription factor Sp1 to induce TGFβ1 expression. In four independent patient cohorts, TGFβ1 expression was increased in the BM of patients with MPN compared with healthy individuals, and the BM of patients with MPN contained a higher frequency of Treg compared with healthy individuals. Together, this study identified an ERK/Sp1/TGFβ1 axis in CALRdel52 MPNs as a mechanism of immunosuppression that can be targeted to elicit T-cell-mediated cytotoxicity. Significance: Targeting the mutant calreticulin/TGFβ1 axis increases T-cell activity and glycolytic capacity, providing the rationale for conducting clinical trials on TGFβ antagonists as an immunotherapeutic strategy in patients with myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Dominik Schmidt
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Cornelia Endres
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Rouven Hoefflin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melissa Zwick
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Nikolaos Karantzelis
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Hans F. Staehle
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Janaki Manoja Vinnakota
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Sandra Duquesne
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Miriam Mozaffari
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Heiko Becker
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Bruce R. Blazar
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alexander Zähringer
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Justus Duyster
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tilman Brummer
- IMMZ, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg
| | - Julian Baumeister
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, and Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Khalid Shoumariyeh
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Juan Li
- Department of Hematology, University of Cambridge, Cambridge, UK
| | - Anthony R. Green
- Department of Hematology, University of Cambridge, Cambridge, UK
| | - Florian H. Heidel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany
- Leibniz Institute on Aging, Fritz-Lipmann Institute, Jena, Germany
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Heike L. Pahl
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Nils Leimkühler
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Germany
| | - Natalie Köhler
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg
| | - Marcelo A. S. de Toledo
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, and 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, and Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Robert Zeiser
- Department of Medicine I - Medical centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg
- Signalling Research Centres BIOSS and CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg
| |
Collapse
|
33
|
Blanco-Sánchez A, Gil-Manso R, de Nicolás R, López-Muñoz N, Colmenares R, Mas R, Sánchez R, Rapado I, Martínez-López J, Díaz RA, Carreño-Tarragona G. Real-World Impact of Deep Targeted Sequencing on Erythrocytosis and Thrombocytosis Diagnosis: A Reference Centre Experience. Cancers (Basel) 2024; 16:3149. [PMID: 39335122 PMCID: PMC11430442 DOI: 10.3390/cancers16183149] [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: 08/18/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Despite advances in diagnosis of erythrocytosis and thrombocytosis due to driver mutation testing, many cases remain classified as "idiopathic". This can be explained by the absence of an evident secondary cause, inconclusive bone marrow biopsy or neglection of family history. Analysis of a broad panel of genes through next-generation sequencing (NGS) could improve diagnostic work-up identifying underlying genetic causes. We reviewed the results of NGS performed in our laboratory and its diagnostic impact on 117 patients with unexplained erythrocytosis and 58 with unexplained thrombocytosis; six patients (5.1%) were diagnosed with polycythaemia vera (PV) and 8 (6.8%) with familial erythrocytosis after NGS testing. Low EPO and a family history seemed to predict a positive result, respectively. However, a greater percentage of patients were ultimately diagnosed with secondary erythrocytosis (36%), remained as idiopathic (28.2%) or were self-limited (15%). The yield of NGS was shown to be slightly higher in patients with thrombocytosis, as 15 (25.9%) were diagnosed with essential thrombocythemia (ET) or familial thrombocytosis after variant detection; previous research has shown similar results, but most of them carried out NGS retrospectively, while the present study exhibits the performance of this test in a real-world setting. Overall, the low rate of variant detection and its poor impact on diagnostic work-up highlights the need for a thorough screening prior to NGS, in order to improve its yield.
Collapse
Affiliation(s)
- Alberto Blanco-Sánchez
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| | - Rodrigo Gil-Manso
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| | - Rodrigo de Nicolás
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| | - Nieves López-Muñoz
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| | - Rafael Colmenares
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| | - Reyes Mas
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| | - Ricardo Sánchez
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| | - Inmaculada Rapado
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| | - Joaquín Martínez-López
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| | - Rosa Ayala Díaz
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| | - Gonzalo Carreño-Tarragona
- Centro Nacional de Investigaciones Oncológicas, Complutense University, 28029 Madrid, Spain; (A.B.-S.); (R.G.-M.); (R.d.N.); (N.L.-M.); (R.C.); (R.M.); (R.S.); (J.M.-L.)
- Hematology Department, Hospital Universitario 12 de Octubre, I + 12, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Oncología, 28029 Madrid, Spain
| |
Collapse
|
34
|
Vadeikienė R, Jakštys B, Laukaitienė D, Šatkauskas S, Juozaitytė E, Ugenskienė R. The Role of Mutated Calreticulin in the Pathogenesis of BCR-ABL1-Negative Myeloproliferative Neoplasms. Int J Mol Sci 2024; 25:9873. [PMID: 39337361 PMCID: PMC11432199 DOI: 10.3390/ijms25189873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs) are characterized by increased proliferation of myeloid lineages in the bone marrow. Calreticulin (CALR) 52 bp deletion and CALR 5 bp insertion have been identified in essential thrombocythemia (ET) and primary myelofibrosis (PMF). There is not much data on the crosstalk between mutated CALR and MPN-related signaling pathways, such as JAK/STAT, PI3K/Akt/mTOR, and Hedgehog. Calreticulin, a multifunctional protein, takes part in many cellular processes. Nevertheless, there is little data on how mutated CALR affects the oxidative stress response and oxidative stress-induced DNA damage, apoptosis, and cell cycle progression. We aimed to investigate the role of the CALR 52 bp deletion and 5 bp insertion in the pathogenesis of MPN, including signaling pathway activation and functional analysis in CALR-mutated cells. Our data indicate that the JAK/STAT and PI3K/Akt/mTOR pathways are activated in CALR-mutated cells, and this activation does not necessarily depend on the CALR and MPL interaction. Moreover, it was found that CALR mutations impair calreticulin function, leading to reduced responses to oxidative stress and DNA damage. It was revealed that the accumulation of G2/M-CALR-mutated cells indicates that oxidative stress-induced DNA damage is difficult to repair. Taken together, this study contributes to a deeper understanding of the specific molecular mechanisms underlying CALR-mutated MPNs.
Collapse
Affiliation(s)
- Roberta Vadeikienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Baltramiejus Jakštys
- Research on Delivery of Medicine and Genes Cluster, Faculty of Natural Sciences, Vytautas Magnus University, LT-44001 Kaunas, Lithuania
| | - Danguolė Laukaitienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Saulius Šatkauskas
- Research on Delivery of Medicine and Genes Cluster, Faculty of Natural Sciences, Vytautas Magnus University, LT-44001 Kaunas, Lithuania
| | - Elona Juozaitytė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Rasa Ugenskienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| |
Collapse
|
35
|
Naskar S, Sriraman N, Sarkar A, Mahajan N, Sarkar K. Tumor antigen presentation and the associated signal transduction during carcinogenesis. Pathol Res Pract 2024; 261:155485. [PMID: 39088877 DOI: 10.1016/j.prp.2024.155485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
Numerous developments have been achieved in the study and treatment of cancer throughout the decades that it has been common. After decades of research, about 100 different kinds of cancer have been found, each with unique subgroups within certain organs. This has significantly expanded our understanding of the illness. A mix of genetic, environmental, and behavioral variables contribute to the complicated and diverse process of cancer formation. Mutations, or changes in the DNA sequence, are crucial to the development of cancer. These mutations have the ability to downregulate the expression and function of Major Histocompatibility Complex class I (MHC I) and MHCII receptors, as well as activate oncogenes and inactivate tumor suppressor genes. Cancer cells use this tactic to avoid being recognized by cytotoxic CD8+T lymphocytes, which causes issues with antigen presentation and processing. This review goes into great length into the PI3K pathway, changes to MHC I, and positive impacts of tsMHC-II on disease-free survival and overall survival and the involvement of dendritic cells (DCs) in different tumor microenvironments. The vital functions that the PI3K pathway and its link to the mTOR pathway are highlighted and difficulties in developing effective cancer targeted therapies and feedback systems has also been mentioned, where resistance mechanisms include RAS-mediated oncogenic changes and active PI3K signalling.
Collapse
Affiliation(s)
- Sohom Naskar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Nawaneetan Sriraman
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Ankita Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Nitika Mahajan
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India.
| |
Collapse
|
36
|
Capitanio D, Calledda FR, Abbonante V, Cattaneo D, Moriggi M, Bartalucci N, Bucelli C, Tosi D, Gianelli U, Vannucchi AM, Iurlo A, Gelfi C, Balduini A, Malara A. Proteomic screening identifies PF4/Cxcl4 as a critical driver of myelofibrosis. Leukemia 2024; 38:1971-1984. [PMID: 39025985 DOI: 10.1038/s41375-024-02354-z] [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: 01/18/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Despite increased understanding of the genomic landscape of Myeloproliferative Neoplasms (MPNs), the pathological mechanisms underlying abnormal megakaryocyte (Mk)-stromal crosstalk and fibrotic progression in MPNs remain unclear. We conducted mass spectrometry-based proteomics on mice with Romiplostim-dependent myelofibrosis to reveal alterations in signaling pathways and protein changes in Mks, platelets, and bone marrow (BM) cells. The chemokine Platelet Factor 4 (PF4)/Cxcl4 was up-regulated in all proteomes and increased in plasma and BM fluids of fibrotic mice. High TPO concentrations sustained in vitro PF4 synthesis and secretion in cultured Mks, while Ruxolitinib restrains the abnormal PF4 expression in vivo. We discovered that PF4 is rapidly internalized by stromal cells through surface glycosaminoglycans (GAGs) to promote myofibroblast differentiation. Cxcl4 gene silencing in Mks mitigated the profibrotic phenotype of stromal cells in TPO-saturated co-culture conditions. Consistently, extensive stromal PF4 uptake and altered GAGs deposition were detected in Romiplostim-treated, JAK2V617F mice and BM biopsies of MPN patients. BM PF4 levels and Mk/platelet CXCL4 expression were elevated in patients, exclusively in overt fibrosis. Finally, pharmacological inhibition of GAGs ameliorated in vivo fibrosis in Romiplostim-treated mice. Thus, our findings highlight the critical role of PF4 in the fibrosis progression of MPNs and substantiate the potential therapeutic strategy of neutralizing PF4-GAGs interaction.
Collapse
Affiliation(s)
- Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | | | - Vittorio Abbonante
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Azienda Ospedaliera-Universitaria Careggi, Florence, Italy
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Delfina Tosi
- Department of Health Sciences, University of Milan, S.C. di Anatomia Patologica, ASST-Santi Paolo e Carlo, Milan, Italy
| | - Umberto Gianelli
- Department of Health Sciences, University of Milan, S.C. di Anatomia Patologica, ASST-Santi Paolo e Carlo, Milan, Italy
| | - Alessandro Maria Vannucchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Azienda Ospedaliera-Universitaria Careggi, Florence, Italy
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | | | - Alessandro Malara
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
37
|
Kristensen DT, Øvlisen AK, Jakobsen LHK, Severinsen MT, Hannig LH, Starklint J, Hilsøe MH, Vallentin AP, Brabrand M, Hasselbalch HC, El-Galaly TC, Roug AS. Metformin use and risk of myeloproliferative neoplasms: a Danish population-based case-control study. Blood Adv 2024; 8:4478-4485. [PMID: 38758071 PMCID: PMC11445222 DOI: 10.1182/bloodadvances.2023012266] [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: 12/05/2023] [Revised: 03/18/2024] [Accepted: 04/11/2024] [Indexed: 05/18/2024] Open
Abstract
ABSTRACT Previous studies have suggested that metformin has beneficial effects beyond its glucose-lowering properties, particularly in terms of its potential as an antineoplastic and cancer-preventive agent. In this study, we aimed to investigate the association between metformin use and the risk of myeloproliferative neoplasms (MPN). We conducted a population-based case-control study using Danish registers. Cases with MPN diagnosed between 2010 and 2018 were identified, and metformin use before the MPN diagnosis was ascertained. We compared metformin use among cases with MPN and an age- and sex-matched control group from the Danish general population to estimate age- and sex-adjusted odds ratios (ORs) and fully adjusted ORs (aORs) for the association between metformin use and risk of MPN. The study population included 3816 cases and 19 080 controls. Overall, 7.0% of cases and 8.2% of controls were categorized as ever-users of metformin, resulting in an OR for MPN of 0.84 (95% confidence interval [CI], 0.73-0.96) and an aOR of 0.70 (95% CI, 0.61-0.81). Long-term metformin use (≥5 years) was more infrequent and comprised 1.1% of cases and 2.0% of controls, resulting in an OR of 0.57 (95% CI, 0.42-0.79) and an aOR of 0.45 (95% CI, 0.33-0.63). A dose-response relationship was observed when cumulative duration of treatment was analyzed, and this was consistent in stratified analyses of sex, age, and MPN subtypes. In conclusion, metformin use was associated with significantly lower odds of an MPN diagnosis, indicating its potential cancer-preventive effect. Given the retrospective design, causality cannot be inferred.
Collapse
Affiliation(s)
- Daniel Tuyet Kristensen
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Andreas Kiesbye Øvlisen
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Lasse Hjort Kyneb Jakobsen
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Marianne Tang Severinsen
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Jørn Starklint
- Department of Internal Medicine, Section for Hematology, Regional Hospital West, Gødstrup, Denmark
| | | | - Anders Pommer Vallentin
- Department of Haematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mette Brabrand
- Department of Haematology, Odense University Hospital, Odense, Denmark
| | | | - Tarec Christoffer El-Galaly
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Haematology Research Unit, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institute, Stockholm, Sweden
| | - Anne Stidsholt Roug
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
38
|
Todor SB, Ichim C, Boicean A, Mihaila RG. Cardiovascular Risk in Philadelphia-Negative Myeloproliferative Neoplasms: Mechanisms and Implications-A Narrative Review. Curr Issues Mol Biol 2024; 46:8407-8423. [PMID: 39194713 DOI: 10.3390/cimb46080496] [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: 07/08/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs), encompassing disorders like polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are characterized by clonal hematopoiesis without the Philadelphia chromosome. The JAK2 V617F mutation is prevalent in PV, ET, and PMF, while mutations in MPL and CALR also play significant roles. These conditions predispose patients to thrombotic events, with PMF exhibiting the lowest survival among MPNs. Chronic inflammation, driven by cytokine release from aberrant leukocytes and platelets, amplifies cardiovascular risk through various mechanisms, including atherosclerosis and vascular remodeling. Additionally, MPN-related complications like pulmonary hypertension and cardiac fibrosis contribute to cardiovascular morbidity and mortality. This review consolidates recent research on MPNs' cardiovascular implications, emphasizing thrombotic risk, chronic inflammation, and vascular stiffness. Understanding these associations is crucial for developing targeted therapies and improving outcomes in MPN patients.
Collapse
Affiliation(s)
- Samuel Bogdan Todor
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Adrian Boicean
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | | |
Collapse
|
39
|
Cattaneo D, Galli N, Bucelli C, Fidanza CA, Bellani V, Artuso S, Bianchi P, Consonni D, Passamonti F, Iurlo A. Red cell distribution width and prognosis in myelofibrosis patients treated with ruxolitinib. Ann Hematol 2024; 103:2787-2795. [PMID: 38864904 DOI: 10.1007/s00277-024-05801-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024]
Abstract
We evaluated RDW in a single-center series of 61 consecutive patients with primary and secondary MF at diagnosis and during treatment with ruxolitinib (RUX) and examined any possible prognostic impact. Elevated RDW values were present in all but 4 patients at diagnosis with a median RDW of 18.9%. RDW was higher in subjects with palpable splenomegaly (p = 0.02), higher ferritin, as well as among those cases who did not receive any cytoreduction before RUX (p = 0.04). Interestingly, higher RDW at diagnosis also correlated with a shorter time from MF diagnosis to RUX start (-4.1 months per one RDW unit; p = 0.03). We observed a modest increase (< 1%) in RDW during the first 6 months of RUX treatment. In a multivariable random-intercept model that considered all time points and contained the covariates time and RUX dose, we also observed a clear decrease in RDW with increasing hemoglobin (Hb) during RUX (slope: -0.4% per g/dL of Hb; p < 0.001). The median RDW at diagnosis of 18.9% was used as a cut-off to identify two subgroups of patients [Group 1: RDW 19.0-25.7%; Group 2: RDW 13.1-18.7%], showing a difference in mortality [Group 1 vs. 2: crude HR 2.88; p = 0.01]. Using continuous RDW at diagnosis, the crude HR was 1.21 per RDW unit (p = 0.002). In a Cox model adjusted for gender, age and Hb at diagnosis, the HR was 1.13 per RDW unit (p = 0.07). RDW may have prognostic significance at MF diagnosis and during RUX, helping in the rapid detection of patients with poor prognosis.
Collapse
Affiliation(s)
- Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Myeloproliferative Syndromes Unit, Via Francesco Sforza 35, Milan, 20122, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Nicole Galli
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Myeloproliferative Syndromes Unit, Via Francesco Sforza 35, Milan, 20122, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Myeloproliferative Syndromes Unit, Via Francesco Sforza 35, Milan, 20122, Italy
| | - Cecilia Anna Fidanza
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Myeloproliferative Syndromes Unit, Via Francesco Sforza 35, Milan, 20122, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Valentina Bellani
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Myeloproliferative Syndromes Unit, Via Francesco Sforza 35, Milan, 20122, Italy
| | - Silvia Artuso
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Myeloproliferative Syndromes Unit, Via Francesco Sforza 35, Milan, 20122, Italy
| | - Paola Bianchi
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Myeloproliferative Syndromes Unit, Via Francesco Sforza 35, Milan, 20122, Italy
| | - Dario Consonni
- Epidemiology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Passamonti
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Myeloproliferative Syndromes Unit, Via Francesco Sforza 35, Milan, 20122, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Myeloproliferative Syndromes Unit, Via Francesco Sforza 35, Milan, 20122, Italy.
| |
Collapse
|
40
|
Kaehler M, von Bubnoff N, Cascorbi I, Gorantla SP. Molecular biomarkers of leukemia: convergence-based drug resistance mechanisms in chronic myeloid leukemia and myeloproliferative neoplasms. Front Pharmacol 2024; 15:1422565. [PMID: 39104388 PMCID: PMC11298451 DOI: 10.3389/fphar.2024.1422565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Leukemia represents a diverse group of hematopoietic neoplasms that can be classified into different subtypes based on the molecular aberration in the affected cell population. Identification of these molecular classification is required to identify specific targeted therapeutic approaches for each leukemic subtype. In general, targeted therapy approaches achieve good responses in some leukemia subgroups, however, resistance against these targeted therapies is common. In this review, we summarize molecular drug resistance biomarkers in targeted therapies in BCR::ABL1-driven chronic myeloid leukemia (CML) and JAK2-driven myeloproliferative neoplasms (MPNs). While acquisition of secondary mutations in the BCR::ABL1 kinase domain is the a common mechanism associated with TKI resistance in CML, in JAK2-driven MPNs secondary mutations in JAK2 are rare. Due to high prevalence and lack of specific therapy approaches in MPNs compared to CML, identification of crucial pathways leading to inhibitor persistence in MPN model is utterly important. In this review, we focus on different alternative signaling pathways activated in both, BCR::ABL1-mediated CML and JAK2-mediated MPNs, by combining data from in vitro and in vivo-studies that could be used as potential biomarkers of drug resistance. In a nutshell, some common similarities, especially activation of PDGFR, Ras, PI3K/Akt signaling pathways, have been demonstrated in both leukemias. In addition, induction of the nucleoprotein YBX1 was shown to be involved in TKI-resistant JAK2-mediated MPN, as well as TKI-resistant CML highlighting deubiquitinating enzymes as potential biomarkers of TKI resistance. Taken together, whole exome sequencing of cell-based or patients-derived samples are highly beneficial to define specific resistance markers. Additionally, this might be helpful for the development of novel diagnostic tools, e.g., liquid biopsy, and novel therapeutic agents, which could be used to overcome TKI resistance in molecularly distinct leukemia subtypes.
Collapse
Affiliation(s)
- Meike Kaehler
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sivahari Prasad Gorantla
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
41
|
Ushijima Y, Naruse S, Ishikawa Y, Kawashima N, Sanada M, Nakashima M, Kim JH, Terakura S, Kihara R, Watamoto K, Nishiyama T, Kitamura K, Matsushita T, Kiyoi H. Initiating-clone analysis in patients with acute myeloid leukemia secondary to essential thrombocythemia. Sci Rep 2024; 14:15906. [PMID: 38987297 PMCID: PMC11237009 DOI: 10.1038/s41598-024-66461-8] [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: 02/12/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Most of essential thrombocythemia (ET) patients have the clone harboring a mutation in one of the JAK2, CALR, or MPL gene, and these clones generally acquire additional mutations at transformation to acute myeloid leukemia (AML). However, the proliferation of triple-negative clones has sometimes been observed at AML transformation. To clarify the clonal evolution of ET to AML, we analyzed paired samples at ET and AML transformation in eight patients. We identified that JAK2-unmutated AML clones proliferated at AML transformation in three patients in whom the JAK2-mutated clone was dominant at ET. In two patients, TET2-mutated, but not JAK2-mutated, clones might be common initiating clones for ET and transformed AML. In a patient with JAK2-mutated ET, SMARCC2, UBR4, and ZNF143, but not JAK2, -mutated clones proliferated at AML transformation. Precise analysis using single-cell sorted CD34+/CD38- fractions suggested that ET clone with JAK2-mutated and AML clone with TP53 mutation was derived from the common clone with these mutations. Although further study is required to clarify the biological significance of SMARCC2, UBR4, and ZNF143 mutations during disease progression of ET and AML transformation, the present results demonstrate the possibility of a common initial clone involved in both ET and transformed AML.
Collapse
Affiliation(s)
- Yoko Ushijima
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Seara Naruse
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yuichi Ishikawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan.
| | - Naomi Kawashima
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Masashi Sanada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Marie Nakashima
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Jeong Hui Kim
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Seitaro Terakura
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Rika Kihara
- Department of Hematology and Oncology, Konan Kosei Hospital, Konan, Japan
- Department of Hematology, Komaki City Hospital, Komaki, Japan
| | - Koichi Watamoto
- Department of Hematology, Komaki City Hospital, Komaki, Japan
| | - Takahiro Nishiyama
- Division of Hematology, Ichinomiya Municipal Hospital, Ichinomiya, Japan
| | - Kunio Kitamura
- Division of Hematology, Ichinomiya Municipal Hospital, Ichinomiya, Japan
| | - Tadashi Matsushita
- Department of Transfusion Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Tsurumai-Cho 65, Showa-Ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
42
|
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
|
43
|
Xiao L, Zhang L, Guo C, Xin Q, Gu X, Jiang C, Wu J. "Find Me" and "Eat Me" signals: tools to drive phagocytic processes for modulating antitumor immunity. Cancer Commun (Lond) 2024; 44:791-832. [PMID: 38923737 PMCID: PMC11260773 DOI: 10.1002/cac2.12579] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Phagocytosis, a vital defense mechanism, involves the recognition and elimination of foreign substances by cells. Phagocytes, such as neutrophils and macrophages, rapidly respond to invaders; macrophages are especially important in later stages of the immune response. They detect "find me" signals to locate apoptotic cells and migrate toward them. Apoptotic cells then send "eat me" signals that are recognized by phagocytes via specific receptors. "Find me" and "eat me" signals can be strategically harnessed to modulate antitumor immunity in support of cancer therapy. These signals, such as calreticulin and phosphatidylserine, mediate potent pro-phagocytic effects, thereby promoting the engulfment of dying cells or their remnants by macrophages, neutrophils, and dendritic cells and inducing tumor cell death. This review summarizes the phagocytic "find me" and "eat me" signals, including their concepts, signaling mechanisms, involved ligands, and functions. Furthermore, we delineate the relationships between "find me" and "eat me" signaling molecules and tumors, especially the roles of these molecules in tumor initiation, progression, diagnosis, and patient prognosis. The interplay of these signals with tumor biology is elucidated, and specific approaches to modulate "find me" and "eat me" signals and enhance antitumor immunity are explored. Additionally, novel therapeutic strategies that combine "find me" and "eat me" signals to better bridge innate and adaptive immunity in the treatment of cancer patients are discussed.
Collapse
Affiliation(s)
- Lingjun Xiao
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Louqian Zhang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Ciliang Guo
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Qilei Xin
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Xiaosong Gu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| |
Collapse
|
44
|
Aguirre LE, Jain A, Ball S, Ali NA, Volpe VO, Tinsley-Vance S, Sallman D, Sweet K, Lancet J, Padron E, Yun S, Kuykendall A, Komrokji R. Triple-Negative Myelofibrosis: Disease Features, Response to Treatment and Outcomes. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:459-467. [PMID: 38548563 DOI: 10.1016/j.clml.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Myelofibrosis is the most aggressive subtype among classical BCR::ABL1 negative myeloproliferative neoplasms. About 90% of cases are driven by constitutive activation of 1 of 3 genes impacting the JAK/STAT pathway: JAK2, CALR, and MPL. Triple-negative myelofibrosis (TN-MF) accounts for only 5%-10% of cases and carries the worst outcomes. Little has been described about this subset of disease. Given the marked heterogeneity surrounding disease biology, clonal architecture, clinical presentation, and poor outcomes in TN-MF, identification of features of interest and assessment of treatment response are areas in need of further investigation. PATIENTS AND METHODS We collected and evaluated baseline clinical and molecular parameters from 626 patients with a diagnosis of myelofibrosis who presented to the H. Lee Moffitt Cancer Center in Tampa (Florida, US) between 2003 and 2021 and compared them based on presence or absence of the three classical phenotypic driver mutations. RESULTS A small proportion of patients (6%) harbored TN-MF which correlated with inferior outcomes, marked by a 4-year reduction in overall survival time compared to the non-TN cohort (mOS 37.4 months vs. 85.7 months; P = .009) and higher rates of leukemic transformation. More pronounced thrombocytopenia and anemia, lower LDH, EPO levels, as well as lower percentage of marrow blasts at baseline were more commonly seen in TN-MF (P < .05). Similarly, patients with TN-MF had higher risk disease per DIPSS+ and GIPSS. Mutations impacting RNA splicing, epigenetic modification and signaling, specifically SRSF2, SETBP1, IDH2, CBL, and GNAS, were more commonly seen among those lacking a classical phenotypic driver. The prevalence of co-mutant ASXL1/SRSF2 clones was significantly higher in TN-MF as was trisomy 8. TN patients had fewer responses (46.2% vs. 63.4%) and shorter duration of response to ruxolitinib. CONCLUSION TN-MF is invariably associated with significantly decreased survival and more aggressive clinical behavior with higher rates of leukemic transformation and shorter duration of response to ruxolitinib. Mutations impacting RNA splicing, epigenetic modification and signaling (SRSF2, SETBP1, IDH2, CBL, and GNAS) are more common in TN-MF, which likely drive its aggressive course and may account for suboptimal responses to JAK inhibition.
Collapse
Affiliation(s)
- Luis E Aguirre
- Department of Medical Oncology, Adult Leukemia Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.
| | - Akriti Jain
- Leukemia and Myeloid Disorders Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Somedeb Ball
- Division of Hematology and Oncology, Vanderbilt Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Najla Al Ali
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Virginia O Volpe
- Department of Medical Oncology, Adult Leukemia Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Sara Tinsley-Vance
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - David Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Kendra Sweet
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Jeffrey Lancet
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Seongseok Yun
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Andrew Kuykendall
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Rami Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL.
| |
Collapse
|
45
|
Hunter AM, Bose P. Advances with janus kinase inhibitors for the treatment of myeloproliferative neoplasms: an update of the literature. Expert Opin Pharmacother 2024; 25:1391-1404. [PMID: 39067001 DOI: 10.1080/14656566.2024.2385729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION The hallmark discovery of hyperactivation of the janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway was a sentinel moment in the history of myeloproliferative neoplasms (MPNs). This finding paved the way for the development of JAK inhibitors, which now represent the foundation of myelofibrosis therapy. With four JAK inhibitors now approved for myelofibrosis, awareness of their clinical efficacy and safety data and recognition of their unique pharmacologic attributes are of critical importance. Additionally, ruxolitinib represents an integral part of the therapeutic arsenal for polycythemia vera. AREAS COVERED This review provides a broad overview of the published literature supporting JAK inhibitor therapy for MPNs. Primarily focusing on myelofibrosis, each of the four available JAK inhibitors is reviewed in detail, including pharmacology, efficacy, and safety data. Failure of JAK inhibitors and future directions in JAK inhibitor therapy are also discussed. EXPERT OPINION JAK inhibitors revolutionized the treatment of MPNs and have dramatically improved patient outcomes. However, data informing selection between currently available JAK inhibitors is limited. These agents are not curative and eventually fail most patients with myelofibrosis. Combining JAK inhibitors with novel targeted agents appears to be the most promising path to further improve outcomes.
Collapse
Affiliation(s)
- Anthony M Hunter
- Department of Hematology and Medical oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
46
|
Huang J, Zhang P, Shen F, Zheng X, Ding Q, Pan Y, Ruan X. Prediction of acute coronary syndrome in patients with myeloproliferative neoplasms. Front Cardiovasc Med 2024; 11:1369701. [PMID: 38984355 PMCID: PMC11231400 DOI: 10.3389/fcvm.2024.1369701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/28/2024] [Indexed: 07/11/2024] Open
Abstract
Background Patients with myeloproliferative neoplasms (MPN) are exposed to a higher risk of cardiovascular disease, especially cardiovascular calcification. The present research aimed to analyze the clinical features and coronary artery calcium score (CACS) in MPN patients, and construct an effective model to predict acute coronary syndrome (ACS) in MPN patients. Materials and methods A total of 175 MPN patients and 175 controls were recruited from the First Affiliated Hospital of Ningbo University. Based on cardiovascular events, the MPN patients were divided into the ACS group and the non-ACS group. Multivariate Cox analysis was completed to explore ACS-related factors. Furthermore, ROC curves were plotted to assess the predictive effect of CACS combined with white blood cells (WBC) and platelet for ACS in MPN patients. Results The MPN group exhibited a higher CACS than the control group (133 vs. 55, P < 0.001). A total of 16 patients developed ACS in 175 MPN patients. Compared with non-ACS groups, significant differences in age, diabetes, smoking history, WBC, percentage of neutrophil, percentage of lymphocyte, neutrophil count, hemoglobin, hematocrit, platelet, lactate dehydrogenase, β 2-microglobulin, and JAK2V617F mutation were observed in the ACS groups. In addition, the CACS in the ACS group was also significantly higher than that in the non-ACS group (374.5 vs. 121, P < 0.001). The multivariable Cox regression analysis identified WBC, platelet, and CACS as independent risk factors for ACS in MPN patients. Finally, ROC curves indicated that WBC, platelet, and CACS have a high predictive value for ACS in MPN patients (AUC = 0.890). Conclusion CACS combined with WBC and platelet might be a promising model for predicting ACS occurrence in MPN patients.
Collapse
Affiliation(s)
- Jingfeng Huang
- Department of Radiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Ping Zhang
- Department of Hematology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Fangjie Shen
- Department of Radiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xiaodong Zheng
- Department of Radiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Qianjiang Ding
- Department of Radiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yuning Pan
- Department of Radiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xinzhong Ruan
- Department of Radiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
47
|
Haage TR, Charakopoulos E, Bhuria V, Baldauf CK, Korthals M, Handschuh J, Müller P, Li J, Harit K, Nishanth G, Frey S, Böttcher M, Fischer KD, Dudeck J, Dudeck A, Lipka DB, Schraven B, Green AR, Müller AJ, Mougiakakos D, Fischer T. Neutrophil-specific expression of JAK2-V617F or CALRmut induces distinct inflammatory profiles in myeloproliferative neoplasia. J Hematol Oncol 2024; 17:43. [PMID: 38853260 PMCID: PMC11163796 DOI: 10.1186/s13045-024-01562-5] [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: 03/22/2024] [Accepted: 05/29/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Neutrophils play a crucial role in inflammation and in the increased thrombotic risk in myeloproliferative neoplasms (MPNs). We have investigated how neutrophil-specific expression of JAK2-V617F or CALRdel re-programs the functions of neutrophils. METHODS Ly6G-Cre JAK2-V617F and Ly6G-Cre CALRdel mice were generated. MPN parameters as blood counts, splenomegaly and bone marrow histology were compared to wild-type mice. Megakaryocyte differentiation was investigated using lineage-negative bone marrow cells upon in vitro incubation with TPO/IL-1β. Cytokine concentrations in serum of mice were determined by Mouse Cytokine Array. IL-1α expression in various hematopoietic cell populations was determined by intracellular FACS analysis. RNA-seq to analyse gene expression of inflammatory cytokines was performed in isolated neutrophils from JAK2-V617F and CALR-mutated mice and patients. Bioenergetics of neutrophils were recorded on a Seahorse extracellular flux analyzer. Cell motility of neutrophils was monitored in vitro (time lapse microscopy), and in vivo (two-photon microscopy) upon creating an inflammatory environment. Cell adhesion to integrins, E-selectin and P-selection was investigated in-vitro. Statistical analysis was carried out using GraphPad Prism. Data are shown as mean ± SEM. Unpaired, two-tailed t-tests were applied. RESULTS Strikingly, neutrophil-specific expression of JAK2-V617F, but not CALRdel, was sufficient to induce pro-inflammatory cytokines including IL-1 in serum of mice. RNA-seq analysis in neutrophils from JAK2-V617F mice and patients revealed a distinct inflammatory chemokine signature which was not expressed in CALR-mutant neutrophils. In addition, IL-1 response genes were significantly enriched in neutrophils of JAK2-V617F patients as compared to CALR-mutant patients. Thus, JAK2-V617F positive neutrophils, but not CALR-mutant neutrophils, are pathogenic drivers of inflammation in MPN. In line with this, expression of JAK2-V617F or CALRdel elicited a significant difference in the metabolic phenotype of neutrophils, suggesting a stronger inflammatory activity of JAK2-V617F cells. Furthermore, JAK2-V617F, but not CALRdel, induced a VLA4 integrin-mediated adhesive phenotype in neutrophils. This resulted in reduced neutrophil migration in vitro and in an inflamed vessel. This mechanism may contribute to the increased thrombotic risk of JAK2-V617F patients compared to CALR-mutant individuals. CONCLUSIONS Taken together, our findings highlight genotype-specific differences in MPN-neutrophils that have implications for the differential pathophysiology of JAK2-V617F versus CALR-mutant disease.
Collapse
Affiliation(s)
- Tobias Ronny Haage
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Emmanouil Charakopoulos
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Vikas Bhuria
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
| | - Conny K Baldauf
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Mark Korthals
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Juliane Handschuh
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Müller
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Juan Li
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, GB, England
| | - Kunjan Harit
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Gopala Nishanth
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Stephanie Frey
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Martin Böttcher
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Jan Dudeck
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Dudeck
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniel B Lipka
- Section of Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
- Faculty of Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Burkhart Schraven
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
| | - Anthony R Green
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, GB, England
| | - Andreas J Müller
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Thomas Fischer
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany.
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
48
|
Bernstein N, Spencer Chapman M, Nyamondo K, Chen Z, Williams N, Mitchell E, Campbell PJ, Cohen RL, Nangalia J. Analysis of somatic mutations in whole blood from 200,618 individuals identifies pervasive positive selection and novel drivers of clonal hematopoiesis. Nat Genet 2024; 56:1147-1155. [PMID: 38744975 PMCID: PMC11176083 DOI: 10.1038/s41588-024-01755-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Human aging is marked by the emergence of a tapestry of clonal expansions in dividing tissues, particularly evident in blood as clonal hematopoiesis (CH). CH, linked to cancer risk and aging-related phenotypes, often stems from somatic mutations in a set of established genes. However, the majority of clones lack known drivers. Here we infer gene-level positive selection in whole blood exomes from 200,618 individuals in UK Biobank. We identify 17 additional genes, ZBTB33, ZNF318, ZNF234, SPRED2, SH2B3, SRCAP, SIK3, SRSF1, CHEK2, CCDC115, CCL22, BAX, YLPM1, MYD88, MTA2, MAGEC3 and IGLL5, under positive selection at a population level, and validate this selection pattern in 10,837 whole genomes from single-cell-derived hematopoietic colonies. Clones with mutations in these genes grow in frequency and size with age, comparable to classical CH drivers. They correlate with heightened risk of infection, death and hematological malignancy, highlighting the significance of these additional genes in the aging process.
Collapse
Affiliation(s)
| | - Michael Spencer Chapman
- Wellcome Sanger Institute, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Kudzai Nyamondo
- Wellcome Sanger Institute, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Zhenghao Chen
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | | | - Emily Mitchell
- Wellcome Sanger Institute, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | | | | | - Jyoti Nangalia
- Wellcome Sanger Institute, Hinxton, UK.
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
| |
Collapse
|
49
|
Sahin Y, Pei J, Baldwin DA, Mansoor N, Koslosky L, Abdelmessieh P, Wang YL, Nejati R, Testa JR. Acute myeloid leukemia with a novel AKAP9::PDGFRA fusion transformed from essential thrombocythemia: A case report and mini review. Leuk Res Rep 2024; 21:100465. [PMID: 38952949 PMCID: PMC11215950 DOI: 10.1016/j.lrr.2024.100465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/07/2024] [Accepted: 05/30/2024] [Indexed: 07/03/2024] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy associated with various combinations of gene mutations, epigenetic abnormalities, and chromosome rearrangement-related gene fusions. Despite the significant degree of heterogeneity in its pathogenesis, many gene fusions and point mutations are recurrent in AML and have been employed in risk stratification over the last several decades. Gene fusions have long been recognized for understanding tumorigenesis and their proven roles in clinical diagnosis and targeted therapies. Advances in DNA sequencing technologies and computational biology have contributed significantly to the detection of known fusion genes as well as for the discovery of novel ones. Several recurring gene fusions in AML have been linked to prognosis, treatment response, and disease progression. In this report, we present a case with a long history of essential thrombocythemia and hallmark CALR mutation transforming to AML characterized by a previously unreported AKAP9::PDGFRA fusion gene. We propose mechanisms by which this fusion may contribute to the pathogenesis of AML and its potential as a molecular target for tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Yavuz Sahin
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jianming Pei
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Don A. Baldwin
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Nashwa Mansoor
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Lori Koslosky
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Peter Abdelmessieh
- Department of Bone Marrow Transplant and Cellular Therapies, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Y. Lynn Wang
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Reza Nejati
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Joseph. R. Testa
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| |
Collapse
|
50
|
Usart M, Hansen N, Stetka J, Almeida Fonseca T, Guy A, Kimmerlin Q, Rai S, Hao-Shen H, Roux J, Dirnhofer S, Skoda RC. The glutaminase inhibitor CB-839 targets metabolic dependencies of JAK2-mutant hematopoiesis in MPN. Blood Adv 2024; 8:2312-2325. [PMID: 38295283 PMCID: PMC11117009 DOI: 10.1182/bloodadvances.2023010950] [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: 06/12/2023] [Revised: 01/04/2024] [Accepted: 01/21/2024] [Indexed: 02/02/2024] Open
Abstract
ABSTRACT Hyperproliferation of myeloid and erythroid cells in myeloproliferative neoplasms (MPN) driven by the JAK2-V617F mutation is associated with altered metabolism. Given the central role of glutamine in anabolic and catabolic pathways, we examined the effects of pharmacologically inhibiting glutaminolysis, that is, the conversion of glutamine (Gln) to glutamate (Glu), using CB-839, a small molecular inhibitor of the enzyme glutaminase (GLS). We show that CB-839 strongly reduced the mitochondrial respiration rate of bone marrow cells from JAK2-V617F mutant (VF) mice, demonstrating a marked dependence of these cells on Gln-derived ATP production. Consistently, in vivo treatment with CB-839 normalized blood glucose levels, reduced splenomegaly and decreased erythrocytosis in VF mice. These effects were more pronounced when CB-839 was combined with the JAK1/2 inhibitor ruxolitinib or the glycolysis inhibitor 3PO, indicating possible synergies when cotargeting different metabolic and oncogenic pathways. Furthermore, we show that the inhibition of glutaminolysis with CB-839 preferentially lowered the proportion of JAK2-mutant hematopoietic stem cells (HSCs). The total number of HSCs was decreased by CB-839, primarily by reducing HSCs in the G1 phase of the cell cycle. CB-839 in combination with ruxolitinib also strongly reduced myelofibrosis at later stages of MPN. In line with the effects shown in mice, proliferation of CD34+ hematopoietic stem and progenitor cells from polycythemia vera patients was inhibited by CB-839 at nanomolar concentrations. These data suggest that inhibiting GLS alone or in combination with inhibitors of glycolysis or JAK2 inhibitors represents an attractive new therapeutic approach to MPN.
Collapse
Affiliation(s)
- Marc Usart
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Nils Hansen
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jan Stetka
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Tiago Almeida Fonseca
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Alexandre Guy
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- UMR1034, Inserm, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France
| | - Quentin Kimmerlin
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Shivam Rai
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Hui Hao-Shen
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Julien Roux
- Bioinformatics core facility, Department of Biomedicine, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Stefan Dirnhofer
- Department of Pathology, University Hospital Basel, Basel, Switzerland
| | - Radek C. Skoda
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|