1
|
Suttantapidok S, Owattanapanich W. Clinical Characteristics, Prognostic Factors, and Thrombotic and Bleeding Outcomes in Philadelphia Chromosome-Negative Myeloproliferative Neoplasms: A Single-Center Cohort Study in Thailand. Cureus 2025; 17:e82141. [PMID: 40231292 PMCID: PMC11994368 DOI: 10.7759/cureus.82141] [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] [Accepted: 04/11/2025] [Indexed: 04/16/2025] Open
Abstract
Background Patients with Philadelphia chromosome-negative myeloproliferative neoplasms (Ph-neg MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), face elevated risks of thrombosis and major bleeding. Because of these potentially severe complications, antiplatelet and anticoagulant therapies are often employed. This study aimed to evaluate thrombotic and bleeding events, identify associated risk factors, assess prognostic risk models, and investigate the effects of antithrombotic therapy in a Thai cohort of patients with Ph-neg MPNs. Methods This single-center cohort study in Thailand enrolled patients with Ph-neg MPNs from 2013 to 2023. Clinical characteristics, prognostic risk models (International Prognostic Scoring System (IPSS), International Prognostic Score for Essential Thrombocythemia (IPSET)-thrombosis, and Dynamic International Prognostic Scoring System (DIPSS)), and arterial and venous thrombotic events and bleeding complications were analyzed using descriptive statistics, logistic regression, and survival analysis. Results Among 173 patients, PV was the most common subtype (n = 111, 64.2%), with a male predominance and a median age of 57 years. Thrombotic events occurred in 36.9% (n = 41) of PV, 21.4% (n = 3) of PMF, and 18.8% (n = 9) of ET cases, with arterial thrombosis, particularly ischemic stroke, being the most frequent. The JAK2 V617F mutation was prevalent in 72.5% (n = 79) of PV, 70.8% (n = 34) of ET, and 50% (n = 5) of PMF patients. Notably, no major bleeding events were observed despite intensive antithrombotic therapy. Multivariable analysis revealed that prior ischemic stroke (OR 22.51, P = 0.007) and dizziness/headache (OR 7.26, P = 0.022) were significant risk factors for thrombosis. Overall survival (OS) varied by disease subtype. PV patients had a five-year OS of 94.9% and a 10-year OS of 87.2%. ET patients demonstrated a five-year OS of 77.4%. PMF patients had the lowest survival, with a five-year OS of 67.7%. Conclusions This cohort study offers important insights into the clinical characteristics and complication profiles of Ph-neg MPNs in a Thai population. Compared with Western cohorts, patients in this study exhibited a higher incidence of thrombotic events, particularly ischemic stroke. These findings emphasize the need to consider regional variations in disease presentation and underscore the value of individualized risk stratification to optimize patient management in diverse clinical settings.
Collapse
Affiliation(s)
| | - Weerapat Owattanapanich
- Department of Medicine, Division of Hematology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, THA
| |
Collapse
|
2
|
Szafraniec HM, Bull F, Higgins JM, Stone HA, Krüger T, Pearce P, Wood DK. Suspension physics govern the multiscale dynamics of blood flow in sickle cell disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.642599. [PMID: 40161781 PMCID: PMC11952557 DOI: 10.1101/2025.03.13.642599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
In diseases from diabetes to malaria, blood dynamics are significantly altered, resulting in poor clinical outcomes. However, the multiscale mechanisms that determine blood flow in the microcirculation in health and disease are undefined, largely owing to the difficulty in directly linking cell properties to whole-blood rheology. Here, we overcome these difficulties by developing a microfluidic platform to measure red blood cell properties and flow dynamics in the same blood samples from donors. We focus on sickle cell disease (SCD), a genetic disorder that causes red blood cells to stiffen in deoxygenated conditions, with disease pathology driven by oxygen-dependent blood rheology. Our linked cell and whole-blood measurements establish that increases in effective resistances in heterogeneous suspensions are driven by increases in the proportion of stiff cells, similar macroscopically to the behavior of rigid-particle suspensions. Furthermore, by combining simulations with spatially resolved measurements of cell dynamics, we show how the spatio-temporal organization of stiff and deformable cells determines blood rheology and drives disease pathophysiology. In the presence of deformable cells, the stiffened cells marginate towards channel walls, increasing effective wall friction. In fully deoxygenated conditions in which all cells are stiffened, significant heterogeneity in cell volume fraction along the direction of flow causes localized jamming, drastically increasing effective viscous flow resistance. Our work defines the relevant suspension physics required to understand pathological blood rheology in SCD and other diseases affecting red blood cell properties. More broadly, we reveal the multiscale processes that determine emergent rheology in heterogeneous particle suspensions.
Collapse
|
3
|
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
|
4
|
Wang YH, Wei CH, Lin CC, Gurnari C, Awada H, Benajiba L, Daltro de Oliveira R, Soret-Dulphy J, Cassinat B, Zucenka A, Mosquera Orgueira A, Yuan CT, Lee SH, Yao CY, Gurashi K, Hou HA, Batta K, Pérez Encinas MM, Chou WC, Maciejewski JP, Wiseman DH, Kiladjian JJ, Tien HF. Synergistic effect of concurrent high molecular risk mutations and lower JAK2 mutant variant allele frequencies on prognosis in patients with myelofibrosis-insights from a multicenter study. Leukemia 2025; 39:144-154. [PMID: 39367172 DOI: 10.1038/s41375-024-02422-4] [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] [Received: 07/29/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/06/2024]
Abstract
In addition to high-molecular risk (HMR) mutations (ASXL1, EZH2, SRSF2, IDH, and U2AF1Q157), lower JAK2V617F variant allele frequencies (VAF) have been demonstrated to be associated with poor prognosis of myelofibrosis (MF) patients. Nevertheless, the relationship between JAK2V617F VAF and HMR mutations remains inconclusive. To address this, we analyzed the mutation status of 54 myeloid neoplasm-relevant genes using targeted next-generation sequencing in 124 MF patients. Three cohorts from multiple international centers were analyzed for external validation. Among JAK2-mutated patients, the presence of HMR mutations drove poor prognosis in patients with lower JAK2V617F VAF but not in those with higher JAK2V617F VAF. Survival analyses showed consistent results across validation cohorts. In multivariable analysis, concurrent HMR and a lower JAK2V617F VAF was identified as an independent adverse prognostic factor for survival, irrespective of age, MIPSS70, MIPSS70 + v2, and GIPSS risk groups. Mutation co-occurrence tests revealed no shared mutational pattern over different cohorts, excluding potential confounding effect from other concurrent mutations. Importantly, the integration of HMR/JAK2V617F VAF (≤50%) status significantly enhanced existing prognostic models, as evidenced by higher c-indexes and time-dependent ROC analyses. Single-cell studies with sequential follow-ups are warranted to decipher the clonal evolution of MF and how it relates to JAK2V617F VAF dynamics.
Collapse
Affiliation(s)
- Yu-Hung Wang
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Epigenetics of Haematopoiesis Laboratory, Division of Cancer Sciences, The University of Manchester, Manchester, UK.
| | - Chao-Hung Wei
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Chien-Chin Lin
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Hussein Awada
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lina Benajiba
- Centre d'Investigations Cliniques, Hospital Saint Louis and University Paris Cité, Paris, France
| | | | - Juliette Soret-Dulphy
- Centre d'Investigations Cliniques, Hospital Saint Louis and University Paris Cité, Paris, France
| | - Bruno Cassinat
- Laboratoire de Biologie Cellulaire, Hospital Saint Louis and University Paris Cité, Paris, France
| | - Andrius Zucenka
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Hematology and Oncology Department, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Adrián Mosquera Orgueira
- Hematology Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Group of Computational Hematology and Genomics, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Chang-Tsu Yuan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Sze-Hwei Lee
- Division of Cellular Therapy, Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan
| | - Chi-Yuan Yao
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kristian Gurashi
- Epigenetics of Haematopoiesis Laboratory, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Hsin-An Hou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kiran Batta
- Epigenetics of Haematopoiesis Laboratory, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | | | - Wen-Chien Chou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Daniel H Wiseman
- Epigenetics of Haematopoiesis Laboratory, Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Jean-Jacques Kiladjian
- Centre d'Investigations Cliniques, Hospital Saint Louis and University Paris Cité, Paris, France
| | - Hwei-Fang Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Internal Medicine, Far-Eastern Memorial Hospital, New Taipei City, Taiwan.
| |
Collapse
|
5
|
Furuya C, Hashimoto Y, Morishita S, Fukuda Y, Inano T, Ochiai T, Shirane S, Edahiro Y, Araki M, Ando M, Komatsu N. A higher JAK2V617F allele burden may be a risk factor for hemorrhagic events in younger patients with polycythemia vera. Hematology 2024; 29:2427905. [PMID: 39545588 DOI: 10.1080/16078454.2024.2427905] [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: 08/08/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
OBJECTIVES Hemorrhagic events are a rare but potentially fatal complication in patients with polycythemia vera (PV). METHODS We analyzed the characteristics of hemorrhagic events in 267 patients with PV. RESULTS A median follow-up of 4.8 years revealed that 23 (8.6%) hemorrhagic events occurred. Significantly more hemorrhagic events occurred in younger patients aged below 60 years (n = 72) than in older patients aged 60 years or above (n = 191) (n = 12 [16.7%] vs. n = 11 [5.8%], respectively, P = 0.012). In univariate analysis among the younger patients, white blood cell (WBC) count ≥ 15 × 109/L (hazard ratio [HR] = 7.746, 95% confidence interval [CI] 2.082-28.830, P = 0.002), palpable splenomegaly (HR = 5.629, 95% CI 1.193-26.550, P = 0.029), and JAK2V617F allele burden ≥ 80% (HR = 22.850, 95% CI 2.885-181.00, P = 0.003) were associated with an increased risk of hemorrhagic events. In multivariate analysis, JAK2V617F allele burden ≥ 80% (HR = 9.394, 95% CI 1.046-84.380, P = 0.046) was a significant risk factor. CONCLUSIONS There is an increased risk of hemorrhagic events after diagnosis in younger PV patients with a high JAK2V617F allele burden, high WBC count or palpable splenomegaly. It is important to consider treatment options that aim to avoid hemorrhagic events by reducing the JAK2V617F allele burden in younger PV patients.
Collapse
Affiliation(s)
- Chiho Furuya
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshinori Hashimoto
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Hematology, Tottori Prefectural Central Hospital, Tottori, Japan
| | - Soji Morishita
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasutaka Fukuda
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tadaaki Inano
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomonori Ochiai
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shuichi Shirane
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoko Edahiro
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Marito Araki
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Miki Ando
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan
- PharmaEssentia Japan KK, Tokyo, Japan
| |
Collapse
|
6
|
Collinson RJ, Wilson L, Boey D, Ng ZY, Mirzai B, Chuah HS, Howman R, Grove CS, Malherbe JAJ, Leahy MF, Linden MD, Fuller KA, Erber WN, Guo BB. Transcription factor 3 is dysregulated in megakaryocytes in myelofibrosis. Platelets 2024; 35:2304173. [PMID: 38303515 DOI: 10.1080/09537104.2024.2304173] [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: 11/15/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024]
Abstract
Transcription factor 3 (TCF3) is a DNA transcription factor that modulates megakaryocyte development. Although abnormal TCF3 expression has been identified in a range of hematological malignancies, to date, it has not been investigated in myelofibrosis (MF). MF is a Philadelphia-negative myeloproliferative neoplasm (MPN) that can arise de novo or progress from essential thrombocythemia [ET] and polycythemia vera [PV] and where dysfunctional megakaryocytes have a role in driving the fibrotic progression. We aimed to examine whether TCF3 is dysregulated in megakaryocytes in MPN, and specifically in MF. We first assessed TCF3 protein expression in megakaryocytes using an immunohistochemical approach analyses and showed that TCF3 was reduced in MF compared with ET and PV. Further, the TCF3-negative megakaryocytes were primarily located near trabecular bone and had the typical "MF-like" morphology as described by the WHO. Genomic analysis of isolated megakaryocytes showed three mutations, all predicted to result in a loss of function, in patients with MF; none were seen in megakaryocytes isolated from ET or PV marrow samples. We then progressed to transcriptomic sequencing of platelets which showed loss of TCF3 in MF. These proteomic, genomic and transcriptomic analyses appear to indicate that TCF3 is downregulated in megakaryocytes in MF. This infers aberrations in megakaryopoiesis occur in this progressive phase of MPN. Further exploration of this pathway could provide insights into TCF3 and the evolution of fibrosis and potentially lead to new preventative therapeutic targets.
Collapse
Affiliation(s)
- Ryan J Collinson
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Lynne Wilson
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Darren Boey
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Zi Yun Ng
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
- Department of Haematology, Royal Perth Hospital, Perth, WA, Australia
| | - Bob Mirzai
- PathWest Laboratory Medicine, Nedlands, WA, Australia
| | - Hun S Chuah
- Department of Haematology, Royal Perth Hospital, Perth, WA, Australia
- PathWest Laboratory Medicine, Nedlands, WA, Australia
- Department of Haematology, Rockingham General Hospital, Rockingham, WA, Australia
| | - Rebecca Howman
- Department of Haematology, Sir Charles Gairdner Hospital Nedlands Australia
| | - Carolyn S Grove
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
- Department of Haematology, Royal Perth Hospital, Perth, WA, Australia
- Department of Haematology, Sir Charles Gairdner Hospital Nedlands Australia
| | | | - Michael F Leahy
- Department of Haematology, Royal Perth Hospital, Perth, WA, Australia
- PathWest Laboratory Medicine, Nedlands, WA, Australia
| | - Matthew D Linden
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Kathryn A Fuller
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Wendy N Erber
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
- PathWest Laboratory Medicine, Nedlands, WA, Australia
| | - Belinda B Guo
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
7
|
Saber S, Abdelhady R, Elhemely MA, Elmorsy EA, Hamad RS, Abdel-Reheim MA, El-Kott AF, AlShehri MA, Morsy K, AlSheri AS, Youssef ME. PU-H71 (NSC 750424): a molecular masterpiece that targets HSP90 in cancer and beyond. Front Pharmacol 2024; 15:1475998. [PMID: 39564119 PMCID: PMC11573589 DOI: 10.3389/fphar.2024.1475998] [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: 08/04/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
Heat shock protein 90 (HSP90) is a pivotal molecular chaperone with multifaceted roles in cellular health and disease. Herein, we explore how HSP90 orchestrates cellular stress responses, particularly through its partnership with heat shock factor 1 (HSF-1). PU-H71, a selective inhibitor of HSP90, demonstrates significant potential in cancer therapy by targeting a wide array of oncogenic pathways. By inducing the degradation of multiple client proteins, PU-H71 disrupts critical signaling pathways such as MAPK, PI3K/Akt, JAK/STAT, EGFR, and mTOR, which are essential for cancer cell survival, proliferation, and metastasis. We examined its impact on combating triple-negative breast cancer and enhancing the effectiveness of carbon-ion beam therapy, offering new avenues for cancer treatment. Furthermore, the dual inhibition of HSP90A and HSP90B1 by PU-H71 proves highly effective in the context of myeloma, providing fresh hope for patients with this challenging malignancy. We delve into its potential to induce apoptosis in B-cell lymphomas that rely on Bcl6 for survival, highlighting its relevance in the realm of hematologic cancers. Shifting our focus to hepatocellular carcinoma, we explore innovative approaches to chemotherapy. Moreover, the current review elucidates the potential capacity of PU-H71 to suppress glial cell activation paving the way for developing novel therapeutic strategies for neuroinflammatory disorders. Additionally, the present report also suggests the promising role of PU-H71 in JAK2-dependent myeloproliferative neoplasms. Eventually, our report sheds more light on the multiple functions of HSP90 protein as well as the potential therapeutic benefit of its selective inhibitor PU-H71 in the context of an array of diseases, laying the foundations for the development of novel therapeutic approaches that could achieve better treatment outcomes.
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Rasha Abdelhady
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Chinese University, Cairo, Egypt
| | - Mai A Elhemely
- School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, Saudi Arabia
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Central Laboratory, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
- Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Mohammed A AlShehri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
- Department of Zoology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Ali S AlSheri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
8
|
Sayed WS, Al-Bayati A, Elzubair LG, Mohamed S, Alharthi M. The Association Between Janus Kinase 2 and Factor V Leiden Mutations and Thrombotic Complications in Patients With Myeloproliferative Disorders: A Study From Saudi Arabia. Cureus 2024; 16:e74401. [PMID: 39723287 PMCID: PMC11669263 DOI: 10.7759/cureus.74401] [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] [Accepted: 11/24/2024] [Indexed: 12/28/2024] Open
Abstract
Background The Janus kinase 2 (JAK2) V617F mutations are related to increased thrombotic risk in patients with myeloproliferative disorders (MPDs). However, little is known about whether inherited thrombophilia represents an additive risk factor in mutated subjects. We addressed the association between combined mutations of JAK2 and factor V Leiden (FVL) and thrombotic complications in Saudi Arabian patients with MPDs. Methods We studied 60 patients with MPDs, 32 with polycythemia vera (PV), 24 with essential thrombocythemia (ET), and four with primary myelofibrosis (PMF). All patients were examined for JAK2 V617F and FVL mutations. Results The study included 50 (83.3%) males and 10 (16.7%) females, with a mean age of 44.23 ± 11.32 years. JAK2 was found positive among all (100%) of the studied patients. Thirty-eight patients out of 60 (63.3%) had thrombotic events. FVL was found positive in 12 (20%) patients. The patients with JAK2 and FVL mutations had a higher incidence of thrombotic events (11/38, 28.9%) than those with JAK2 but without FVL mutations (1/22, 4.5%). The relative risk ratios for increased risk for having thrombotic events were 2.1 (95% confidence interval (95% CI): 1.2-3.8, p=0.03) and 4.3 (95% CI: 2.1-9.5, p<0.001) for patients with JAK2 mutations alone, and those with both JAK2 and FVL mutations, respectively. Conclusions In the present study of patients with MPDs from Saudi Arabia, JAK2 mutations were found among all the studied patients, and FVL mutations were encountered in 20% of patients. The patients with both JAK2 and FVL mutations had a higher incidence of thrombotic events than those with JAK2 but without FVL mutations. The relative risk ratios for increased risk for thrombotic events among patients with MPDs were 2.1 and 4.3 for patients with JAK2 mutations alone and those with JAK2 and FVL mutations, respectively. Further larger prospective studies are warranted.
Collapse
Affiliation(s)
- Wafaa S Sayed
- Clinical Pathology, Aswan University Hospital, Aswan, EGY
- Clinical Pathology, Khamis Mushait General Hospital, Khamis Mushait, SAU
| | - Aws Al-Bayati
- Clinical Hematology, Armed Forces Hospital - Southern Region, Khamis Mushait, SAU
| | - Lina G Elzubair
- Haematopathology, Armed Forces Hospital - Southern Region, Khamis Mushait, SAU
| | - Sherif Mohamed
- Chest Diseases and Tuberculosis, Faculty of Medicine, Assiut University, Assiut, EGY
- Internal Medicine, Prince Sultan Bin Abdulaziz Humanitarian City, Riyadh, SAU
| | - Mueed Alharthi
- Clinical Hematology, Khamis Mushait General Hospital, Khamis Mushait, SAU
| |
Collapse
|
9
|
Holmström MO, Ruders JH, Riley CH, Larsen MK, Grauslund JH, Kjær L, Skov V, Ellervik C, Guo BB, Linden M, Hasselbalch HC, Andersen MH. The CALR mutations enhance the expression of the immunosuppressive proteins GARP and LAP on peripheral blood lymphocytes through increased binding of activated platelets. Br J Haematol 2024; 205:1417-1429. [PMID: 39161981 DOI: 10.1111/bjh.19711] [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: 07/02/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024]
Abstract
Recently, an antibody which inhibits the glycoprotein A repetitions predominant (GARP)-mediated release of active transforming growth factor beta (TGFβ) from the TGFβ propeptide latency-associated peptide (LAP) showed preclinical activity in a murine model of the chronic myeloproliferative neoplasms (MPN). Consequently, we investigated the expression of the immunosuppressive molecules LAP and GARP on peripheral blood lymphocytes from 56 MPN patients and 11 healthy donors (HD). We found that lymphocytes from patients with MPN express higher levels of LAP and GARP with no strong differences found between the different MPN diagnoses. The impact of clinical parameters on the expression of LAP and GARP by lymphocytes showed that patients with calreticulin (CALR)mut MPN have increased expression compared with HD and patients with the Januskinase2 (JAK2) mutation. The fraction of lymphocytes bound to activated platelets (aPLT) strongly correlate to LAP and GARP expression suggesting that it is not the lymphocytes themselves but aPLT, which confer the increased expression of GARP and LAP on MPN patient lymphocytes. Notably, no differences in neither platelet counts nor anti-thrombotic therapy was identified between patients with JAK2- and CALRmut patients. Analysis of platelet gene expression failed to identify differences in expression of relevant genes between JAK2- and CALRmut patients.
Collapse
Affiliation(s)
- Morten Orebo Holmström
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
| | - Josephine Hallundbæk Ruders
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
| | | | | | - Jacob Handlos Grauslund
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
| | - Lasse Kjær
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Vibe Skov
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Department of Clinical Biochemistry, Zealand University Hospital, Koege, Denmark
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Belinda B Guo
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Matthew Linden
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | | | - Mads Hald Andersen
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Tekbaş A, Schilling K, Fahrner R, Morath O, Malessa C, Bauschke A, Settmacher U, Rauchfuß F. Liver Transplantation for Budd-Chiari Syndrome From Myeloproliferative Neoplasms - Management and Long-Term Results. Transplant Proc 2024; 56:1759-1765. [PMID: 39237386 DOI: 10.1016/j.transproceed.2024.08.040] [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: 04/08/2024] [Accepted: 08/24/2024] [Indexed: 09/07/2024]
Abstract
Myeloproliferative neoplasms can cause primary Budd-Chiari-Syndrome with acute or chronic liver failure necessitating liver transplantation. However, preventing the recurrence remains challenging and the need for post-transplant anticoagulant and cytoreductive treatment is not sufficiently clear. We analyzed the treatment regimens for all patients who presented to our department with PBCS from MPN between 2004 and 2021. Eight patients underwent liver transplantation - 6 of them due to an acute liver failure. Post-transplant, all patients received anticoagulant and 7 patients cytoreductive medication. The mean survival after transplantation was 13.25 years. Liver transplantation shows favorable long-term outcome when combined with post-transplant anticoagulant and cytoreductive treatment.
Collapse
Affiliation(s)
- Aysun Tekbaş
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Friedrich Schiller University, Jena, Germany; Research Programme "Advanced Clinician Scientist Programme" by the Interdisciplinary Center of Clinical Research of the Medical Faculty Jena, Jena University Hospital, Friedrich Schiller University, Jena, Germany.
| | - Kristina Schilling
- Department of Hematology and Medical Oncology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - René Fahrner
- Spital Thun, Department of General and Visceral Surgery, Thun, Switzerland
| | - Olga Morath
- Department of Hematology and Medical Oncology, Jena University Hospital, Friedrich Schiller University, Jena, Germany; Research Programme "DFG Clinician Scientist Programme Organ Age" by the Interdisciplinary Center of Clinical Research of the Medical Faculty Jena, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Christina Malessa
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Astrid Bauschke
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Utz Settmacher
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Falk Rauchfuß
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
11
|
Parmar K, Kundu R, Maiti A, Ball S. Updates in biology, classification, and management of acute myeloid leukemia with antecedent hematologic disorder and therapy related acute myeloid leukemia. Leuk Res 2024; 144:107546. [PMID: 38986173 DOI: 10.1016/j.leukres.2024.107546] [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: 01/20/2024] [Revised: 06/03/2024] [Accepted: 06/21/2024] [Indexed: 07/12/2024]
Abstract
Acute myeloid leukemia with antecedent hematologic disorder (AHD-AML) and therapy related AML (t-AML) constitute a heterogenous disease with inferior outcomes. It is often characterized by high-risk cytogenetic and molecular alterations associated with AHD or prior cancer therapy. Historically, the standard of care treatment has been intensive induction with "7 + 3", with an improved overall response rate and survival with CPX-351. Results from large registry-based studies suggested that allogeneic hematopoietic stem cell transplant is preferable to consolidation chemotherapy alone for achieving long-term survival in patients with AHD-AML. Prevalence of high-risk genetic features and advanced age and comorbidities in patients make AHD-AML and t-AML clinically challenging subgroups to treat with intensive approaches. Recent reports on less intensive treatment options, particularly the hypomethylating agent-venetoclax combination, have shown encouraging response rates in these patients. However, emerging resistance mechanisms compromise duration of response and overall survival. Several novel agents targeting apoptotic machinery, signaling pathways, and immune checkpoints are under clinical investigation, with an aim to truly improve overall outcomes in this subgroup. We reviewed updates in biology, classification, and clinical data comparing safety and efficacy of intensive and less intensive treatment options, and summarized ongoing studies with promising novel therapies in AHD-AML and t-AML.
Collapse
Affiliation(s)
- Kanak Parmar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Rupayan Kundu
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Somedeb Ball
- Division of Hematology and Oncology, Vanderbilt University School of Medicine and Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
| |
Collapse
|
12
|
Rubic M, Lee L, Stevenson W. Clinical significance of low level JAK2 V617F in peripheral blood testing. Pathology 2024; 56:726-728. [PMID: 38395679 DOI: 10.1016/j.pathol.2023.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 10/12/2023] [Accepted: 10/28/2023] [Indexed: 02/25/2024]
Affiliation(s)
- Matthew Rubic
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia.
| | - Linda Lee
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - William Stevenson
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, St Leonards, NSW, Australia
| |
Collapse
|
13
|
Xiong H, Liao M, Zhang H, Li Y, Bai J, Zhang J, Li L, Zhang L. DARS expression in BCR/ABL1-negative myeloproliferative neoplasms and its association with the immune microenvironment. Sci Rep 2024; 14:16711. [PMID: 39030308 PMCID: PMC11271514 DOI: 10.1038/s41598-024-67067-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: 04/15/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
DARS, encoding for aspartyl-tRNA synthetase, is implicated in the pathogenesis of various cancers, including renal cell carcinoma, glioblastoma, colon cancer, and gastric cancer. Its role in BCR/ABL1-negative myeloproliferative neoplasms (MPNs), however, remains unexplored. This study aimed to elucidate the expression of DARS in patients with MPNs (PV 23, ET 19, PMF 16) through immunohistochemical analysis and to examine the profiles of circulating immune cells and cytokines using flow cytometry. Our findings indicate a significant overexpression of DARS in all MPNs subtypes at the protein level compared to controls (P < 0.05). Notably, elevated DARS expression was linked to splenomegaly in MPNs patients. The expression of DARS showed a negative correlation with CD4+ T cells (R = - 0.451, P = 0.0004) and CD4+ T/CD8+ T cell ratio (R = - 0.3758, P = 0.0040), as well as with CD68+ tumor-associated macrophages (R = 0.4037, P = 0.0017). Conversely, it was positively correlated with IL-2 (R = 0.5419, P < 0.001), IL-5 (R = 0.3161, P = 0.0166), IL-6 (R = 0.2992, P = 0.0238), and IFN-γ (R = 0.3873, P = 0.0029). These findings underscore a significant association between DARS expression in MPNs patients and specific clinical characteristics, as well as immune cell composition. Further investigation into the interplay between DARS and the immune microenvironment in MPNs could shed light on the underlying mechanisms of MPNs pathogenesis and immune dysregulation.
Collapse
Affiliation(s)
- Hao Xiong
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Minjing Liao
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Huitao Zhang
- Department of General Practice, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yanhong Li
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jun Bai
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jinping Zhang
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China.
| | - Liansheng Zhang
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
14
|
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
|
15
|
Celik H, Challen GA. Enhanced Molecular Response in Myeloproliferative Neoplasms with Complete JAK2V617F Inhibition. Cancer Discov 2024; 14:701-703. [PMID: 38690601 DOI: 10.1158/2159-8290.cd-23-1522] [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: 05/02/2024]
Abstract
SUMMARY Dunbar, Bowman, and colleagues present here a novel genetic mouse model with inducible and reversible expression of the JAK2V617F mutation in the endogenous locus. Results from this study clearly demonstrate an absolute requirement for myeloproliferative neoplasm-initiating cells for this mutation in their survival and imply that more efficacious inhibitors could be curative for these patients even in the setting of additional cooperating mutations. See related article by Dunbar et al., p. 737 (8).
Collapse
Affiliation(s)
- Hamza Celik
- Incyte Research Institute, Wilmington, Delaware
| | - Grant A Challen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
16
|
Kramer F, Mullally A. Antibody targeting of mutant calreticulin in myeloproliferative neoplasms. J Cell Mol Med 2024; 28:e17896. [PMID: 37551061 PMCID: PMC10902560 DOI: 10.1111/jcmm.17896] [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: 05/27/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023] Open
Abstract
Mutations in calreticulin are one of the key disease-initiating mutations in myeloproliferative neoplasms (MPN). In MPN, mutant calreticulin translates with a novel C-terminus that leads to aberrant binding to the extracellular domain of the thrombopoietin receptor, MPL. This cell surface neoantigen has become an attractive target for immunological intervention. Here, we summarize recent advances in the development of mutant calreticulin targeting antibodies as a novel therapeutic approach in MPN.
Collapse
Affiliation(s)
- Frederike Kramer
- Division of Hematology, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Broad InstituteCambridgeMassachusettsUSA
| |
Collapse
|
17
|
Toma MM, Karami A, Nieborowska-Skorska M, Chirtala KN, Pepek M, Hadzijusufovic E, Stoklosa T, Valent P, Skorski T. Clonal medicine targeting DNA damage response eradicates leukemia. Leukemia 2024; 38:671-675. [PMID: 38228681 PMCID: PMC10912018 DOI: 10.1038/s41375-024-02138-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Affiliation(s)
- Monika M Toma
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Adam Karami
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Margaret Nieborowska-Skorska
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Kumaraswamy Naidu Chirtala
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Monika Pepek
- Department of Tumor Biology and Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Emir Hadzijusufovic
- Ludwig Boltzmann Institute for Hematology and Oncology and Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Department for Companion Animals & Horses, Clinic for Internal Medicine and Infectious Diseases, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Tomasz Stoklosa
- Department of Tumor Biology and Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Peter Valent
- Ludwig Boltzmann Institute for Hematology and Oncology and Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Tomasz Skorski
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA.
| |
Collapse
|
18
|
Gou P, Liu D, Ganesan S, Lauret E, Maslah N, Parietti V, Zhang W, Meignin V, Kiladjian JJ, Cassinat B, Giraudier S. Genomic and functional impact of Trp53 inactivation in JAK2V617F myeloproliferative neoplasms. Blood Cancer J 2024; 14:1. [PMID: 38177095 PMCID: PMC10766605 DOI: 10.1038/s41408-023-00969-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/26/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
Classical myeloproliferative neoplasms (MPNs) are characterized by the proliferation of myeloid cells and the risk of transformation into myelofibrosis or acute myeloid leukemia (AML) and TP53 mutations in MPN patients are linked to AML. However, JAK2V617F has been reported to impact the TP53 response to DNA damage, suggesting potential overlapping role of TP53 inactivation in MPN. We established a mouse model showing that JAK2V617F/Vav-Cre/Trp53-/- mice displayed a similar phenotype to JAK2V617F/Vav-Cre mice, but their proliferation was outcompeted in competitive grafts. RNA-Seq revealed that half of the genes affected by JAK2V617F were affected by p53-inactivation, including the interferon pathway. To validate this finding, mice were repopulated with a mixture of wild-type and JAK2V617F (or JAK2V617F/Vav-Cre/Trp53-/-) cells and treated with pegylated interferonα. JAK2V617F-reconstituted mice entered complete hematological remission, while JAK2V617F/Vav-Cre /Trp53-/--reconstituted mice did not, confirming that p53 loss induced interferon-α resistance. KEGG and Gene Ontology analyses of common deregulated genes showed that these genes were mainly implicated in cytokine response, proliferation, and leukemia evolution, illustrating that in this mouse model, the development of MPN is not affected by TP53 inactivation. Taken together, our results show that many genetic modifications induced by JAK2V617F are influenced by TP53, the MPN phenotype may not be. Trp53 loss alone is insufficient to induce rapid leukemic transformation in steady-state hematopoiesis in JAK2V617F MPN, and Trp53 loss may contribute to interferon resistance in MPN.
Collapse
Affiliation(s)
- Panhong Gou
- Inserm UMR-S 1131, Hôpital Saint-Louis, Paris, France
- Université de Paris Cité, Paris, France
| | - Duanya Liu
- Inserm UMR-S 1131, Hôpital Saint-Louis, Paris, France
- Université de Paris Cité, Paris, France
| | | | - Evelyne Lauret
- Université de Paris, Institut Cochin, Inserm U1016, CNRS UMR 8104, Paris, France
| | - Nabih Maslah
- Inserm UMR-S 1131, Hôpital Saint-Louis, Paris, France
- Université de Paris Cité, Paris, France
- Service de Biologie Cellulaire, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Veronique Parietti
- Université de Paris Cité, Paris, France
- INSERM/CNRS US53/UAR2030, Institut de Recherche Saint-Louis, Paris, France
| | | | - Véronique Meignin
- Université de Paris Cité, Paris, France
- Histo-pathological Department, Hôpital Saint-Louis, Paris, France
| | - Jean-Jacques Kiladjian
- Inserm UMR-S 1131, Hôpital Saint-Louis, Paris, France
- Université de Paris Cité, Paris, France
- Centre Investigations Cliniques, Hôpital Saint-Louis, Paris, France
| | - Bruno Cassinat
- Inserm UMR-S 1131, Hôpital Saint-Louis, Paris, France
- Service de Biologie Cellulaire, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Stephane Giraudier
- Inserm UMR-S 1131, Hôpital Saint-Louis, Paris, France.
- Université de Paris Cité, Paris, France.
- Service de Biologie Cellulaire, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France.
| |
Collapse
|
19
|
Kunpara JA, Darji BP, Patel HA, Patwari D, Patel SS, Darji PI. Focal Crescentic Glomerulonephritis Superimposed on Myeloproliferative Disease-Related Glomerulopathy in a Case of Myelofibrosis. Indian J Nephrol 2024; 34:64-66. [PMID: 38645909 PMCID: PMC11003585 DOI: 10.4103/ijn.ijn_218_22] [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: 06/23/2022] [Accepted: 08/21/2022] [Indexed: 04/23/2024] Open
Abstract
Proliferative glomerulonephritis in myelofibrosis is a very rare. Mesangial proliferation and sclerosis with changes of chronic thrombotic microangiopathy have been reported, but pauci-immune focal crescentic glomerulonephritis has not been described so far. Herein, we present a 68-year-old male who was a known case of myelofibrosis and presented with rapidly progressive glomerulonephritis and nephrotic range proteinuria. He was diagnosed as anti-neutrophil cytoplasmic antibody (ANCA)-negative focal crescentic glomerulonephritis, and he responded well to a course of intravenous methylprednisolone and cyclophosphamide. Pauci-immune focal crescentic glomerulonephritis may occur in myelofibrosis without ANCA and may be related to unknown pathogenetic mechanisms in myeloproliferative disorders or suggest any superimposed pathology that might respond well to immunosuppressants.
Collapse
Affiliation(s)
| | | | | | - Devang Patwari
- Department of Nephrology, Zydus Hospitals, Gujarat, India
| | | | | |
Collapse
|
20
|
de Castro FA, Mehdipour P, Chakravarthy A, Ettayebi I, Loo Yau H, Medina TS, Marhon SA, de Almeida FC, Bianco TM, Arruda AGF, Devlin R, de Figueiredo-Pontes LL, Chahud F, da Costa Cacemiro M, Minden MD, Gupta V, De Carvalho DD. Ratio of stemness to interferon signalling as a biomarker and therapeutic target of myeloproliferative neoplasm progression to acute myeloid leukaemia. Br J Haematol 2024; 204:206-220. [PMID: 37726227 DOI: 10.1111/bjh.19107] [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: 05/02/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/21/2023]
Abstract
Progression to aggressive secondary acute myeloid leukaemia (sAML) poses a significant challenge in the management of myeloproliferative neoplasms (MPNs). Since the physiopathology of MPN is closely linked to the activation of interferon (IFN) signalling and that AML initiation and aggressiveness is driven by leukaemia stem cells (LSCs), we investigated these pathways in MPN to sAML progression. We found that high IFN signalling correlated with low LSC signalling in MPN and AML samples, while MPN progression and AML transformation were characterized by decreased IFN signalling and increased LSC signature. A high LSC to IFN expression ratio in MPN patients was associated with adverse clinical prognosis and higher colony forming potential. Moreover, treatment with hypomethylating agents (HMAs) activates the IFN signalling pathway in MPN cells by inducing a viral mimicry response. This response is characterized by double-stranded RNA (dsRNA) formation and MDA5/RIG-I activation. The HMA-induced IFN response leads to a reduction in LSC signature, resulting in decreased stemness. These findings reveal the frequent evasion of viral mimicry during MPN-to-sAML progression, establish the LSC-to-IFN expression ratio as a progression biomarker, and suggests that HMAs treatment can lead to haematological response in murine models by re-activating dsRNA-associated IFN signalling.
Collapse
Affiliation(s)
- Fabíola Attié de Castro
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Parinaz Mehdipour
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| | - Ankur Chakravarthy
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ilias Ettayebi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Helen Loo Yau
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Tiago Silva Medina
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Translational Immuno-Oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Sajid A Marhon
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Felipe Campos de Almeida
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto de Investigação em Imunologia, Institutos Nacionais de Ciência e Tecnologia (INCT-iii), Salvador, Brazil
| | - Thiago Mantello Bianco
- Hematology Division, Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Andrea G F Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Rebecca Devlin
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Lorena Lobo de Figueiredo-Pontes
- Hematology Division, Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Chahud
- Department of Pathology and Forensic Medicine, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maira da Costa Cacemiro
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Vikas Gupta
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Parducci NS, Garnique ADMB, Lima K, Carlos JAEG, Fonseca NP, de Miranda LBL, de Almeida BO, Rego EM, Traina F, Machado-Neto JA. Antineoplastic effects of pharmacological inhibitors of aurora kinases in CSF3R T618I-driven cells. Blood Cells Mol Dis 2024; 104:102799. [PMID: 37839173 DOI: 10.1016/j.bcmd.2023.102799] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/01/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023]
Abstract
Myeloproliferative neoplasms (MPN) are consolidated as a relevant group of diseases derived from the malfunction of the hematopoiesis process and have as a particular attribute the increased proliferation of myeloid lineage. Among these, chronic neutrophilic leukemia (CNL) is distinguished, caused by the T618I mutation of the CSF3R gene, a trait that generates ligand-independent receptor activation and downstream JAK2/STAT signaling. Previous studies reported that mutations in BCR::ABL1 and JAK2V617F increased the expression of the aurora kinase A (AURKA) and B (AURKB) in Ba/F3 cells and their pharmacological inhibition displays antineoplastic effects in human BCR::ABL1 and JAK2V617F positive cells. Delimiting the current scenario, aspects related to the AURKA and AURKB as a potential target in CSF3RT618I-driven models is little known. In the present study, the cellular and molecular effects of pharmacological inhibitors of aurora kinases, such as aurora A inhibitor I, AZD1152-HQPA, and reversine, were evaluated in Ba/F3 expressing the CSF3RT618I mutation. AZD1152-HQPA and reversine demonstrated antineoplastic potential, causing a decrease in cell viability, clonogenicity, and proliferative capacity. At molecular levels, all inhibitors reduced histone H3 phosphorylation, aurora A inhibitor I and reversine reduced STAT5 phosphorylation, and AZD1152-HQPA and reversine induced PARP1 cleavage and γH2AX expression. Reversine more efficiently modulated genes associated with cell cycle and apoptosis compared to other drugs. In summary, our findings shed new insights into the use of AURKB inhibitors in the context of CNL.
Collapse
Affiliation(s)
- Natália Sudan Parducci
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Keli Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | | | - Natasha Peixoto Fonseca
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; Center for Cell Based Therapy, São Paulo Research Foundation, Ribeirão Preto, SP, Brazil
| | | | - Bruna Oliveira de Almeida
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eduardo Magalhães Rego
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil; Center for Cell Based Therapy, São Paulo Research Foundation, Ribeirão Preto, SP, Brazil
| | - Fabiola Traina
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; Center for Cell Based Therapy, São Paulo Research Foundation, Ribeirão Preto, SP, Brazil
| | - João Agostinho Machado-Neto
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
22
|
Torres DG, Barbosa Alves EV, Araújo de Sousa M, Laranjeira WH, Paes J, Alves E, Canté D, Costa AG, Malheiro A, Abreu R, Nascimento L, Fraiji NA, Silva GA, Mourão LPDS, Tarragô AM. Molecular landscape of the JAK2 gene in chronic myeloproliferative neoplasm patients from the state of Amazonas, Brazil. Biomed Rep 2023; 19:98. [PMID: 37954635 PMCID: PMC10633817 DOI: 10.3892/br.2023.1680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/22/2023] [Indexed: 11/14/2023] Open
Abstract
JAK2V617F (dbSNP: rs77375493) is the most frequent and most-studied variant in BCR::ABL1 negative myeloproliferative neoplasms and in the JAK2 gene. The present study aimed to molecularly characterize variants in the complete coding region of the JAK2 gene in patients with BCR::ABL1 negative chronic myeloproliferative neoplasms. The study included 97 patients with BCR::ABL1 negative myeloproliferative neoplasms, including polycythemia vera (n=38), essential thrombocythemia (n=55), and myelofibrosis (n=04). Molecular evaluation was performed using conventional PCR and Sanger sequencing to detect variants in the complete coding region of the JAK2 gene. The presence of missense variants in the JAK2 gene including rs907414891, rs2230723, rs77375493 (JAK2V617F), and rs41316003 were identified. The coexistence of variants was detected in polycythemia vera and essential thrombocythemia. Thus, individuals with high JAK2V617F variant allele frequency (≥50% VAF) presented more thrombo-hemorrhagic events and manifestations of splenomegaly compared with those with low JAK2V617F variant allele frequency (<50% VAF). In conclusion, individuals with BCR::ABL1 negative neoplasms can display >1 variant in the JAK2 gene, especially rs2230722, rs2230724, and rs77375493 variants, and those with high JAK2V617F VAF show alterations in the clinical-laboratory profile compared with those with low JAK2V617F VAF.
Collapse
Affiliation(s)
- Dania G. Torres
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
- Molecular Biology Center, University of Central America, Managua 14003, Nicaragua
| | - Emanuela V. Barbosa Alves
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Miliane Araújo de Sousa
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Wanessa H. Laranjeira
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Jhemerson Paes
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Erycka Alves
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Deborah Canté
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Allyson G. Costa
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
- Post-graduate Program in Basic and Applied Immunology, Federal University of Amazonas, Manaus, Amazonas State 69067-005, Brazil
- Manaus School of Nursing, Federal University of Amazonas, Manaus, Amazonas State 69057-070, Brazil
- Amazon Genomic Health Surveillance Network Coordination, Manaus, Amazonas State 69040-010, Brazil
| | - Adriana Malheiro
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
- Post-graduate Program in Basic and Applied Immunology, Federal University of Amazonas, Manaus, Amazonas State 69067-005, Brazil
- Amazon Genomic Health Surveillance Network Coordination, Manaus, Amazonas State 69040-010, Brazil
| | - Rosângela Abreu
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Leny Nascimento
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Nelson A. Fraiji
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - George A.V. Silva
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Amazon Genomic Health Surveillance Network Coordination, Manaus, Amazonas State 69040-010, Brazil
- Leonidas and Maria Deane Institute, Oswaldo Cruz Foundation, Manaus, Amazonas State 69027-070, Brazil
| | - Lucivana P. de Souza Mourão
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Superior School of Health Sciences, Amazonas State University, Manaus, Amazonas State 69065-001, Brazil
| | - Andréa M. Tarragô
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
- Post-graduate Program in Basic and Applied Immunology, Federal University of Amazonas, Manaus, Amazonas State 69067-005, Brazil
- Amazon Genomic Health Surveillance Network Coordination, Manaus, Amazonas State 69040-010, Brazil
| |
Collapse
|
23
|
Skov V, Thomassen M, Kjaer L, Larsen MK, Knudsen TA, Ellervik C, Kruse TA, Hasselbalch HC. Whole blood transcriptional profiling reveals highly deregulated atherosclerosis genes in Philadelphia-chromosome negative myeloproliferative neoplasms. Eur J Haematol 2023; 111:805-814. [PMID: 37640394 DOI: 10.1111/ejh.14081] [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: 06/26/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND The Philadelphia-negative chronic myeloproliferative neoplasms (MPNs) are associated with a huge comorbidity burden, including an increased risk of cardiovascular diseases. Recently, chronic inflammation has been suggested to be the driving force for clonal evolution and disease progression in MPN but also potentially having an impact upon the development of accelerated (premature) atherosclerosis. OBJECTIVES Since chronic inflammation, atherosclerosis, and atherothrombosis are prevalent in MPNs and we have previously shown oxidative stress genes to be markedly upregulated in MPNs, we hypothesized that genes linked to development of atherosclerosis might be highly deregulated as well. METHODS Using whole blood gene expression profiling in patients with essential thrombocythemia (ET; n = 19), polycythemia vera (PV; n = 41), or primary myelofibrosis (PMF; n = 9), we herein for the first time report aberrant expression of several atherosclerosis genes. RESULTS Of 84 atherosclerosis genes, 45, 56, and 46 genes were deregulated in patients with ET, PV, or PMF, respectively. Furthermore, BCL2L1, MMP1, PDGFA, PTGS1, and THBS4 were progressively significantly upregulated and BCL2 progressively significantly downregulated from ET over PV to PMF (all FDR <0.05). CONCLUSIONS We have for the first time shown massive deregulation of atherosclerosis genes in MPNs, likely reflecting the inflammatory state in MPNs in association with in vivo activation of leukocytes, platelets, and endothelial cells being deeply involved in the atherosclerotic process.
Collapse
Affiliation(s)
- Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Lasse Kjaer
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | | | - Trine A Knudsen
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Torben A Kruse
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | | |
Collapse
|
24
|
Kim HJ, Jung M, Lim BJ, Cheong JW, Han SH. Primary myelofibrosis-related glomerulopathy with renal extramedullary hematopoiesis. J Nephrol 2023; 36:2387-2390. [PMID: 37067698 DOI: 10.1007/s40620-023-01621-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/03/2023] [Indexed: 04/18/2023]
Affiliation(s)
- Hyo Jeong Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Institute of Kidney Disease Research, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Minsun Jung
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Beom Jin Lim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - June-Won Cheong
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Hyeok Han
- Department of Internal Medicine, Yonsei University College of Medicine, Institute of Kidney Disease Research, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
25
|
Holmström MO, Andersen M, Traynor S, Ahmad SM, Lisle TL, Handlos Grauslund J, Skov V, Kjær L, Ottesen JT, Gjerstorff MF, Hasselbalch HC, Andersen MH. Therapeutic cancer vaccination against mutant calreticulin in myeloproliferative neoplasms induces expansion of specific T cells in the periphery but specific T cells fail to enrich in the bone marrow. Front Immunol 2023; 14:1240678. [PMID: 37662956 PMCID: PMC10470021 DOI: 10.3389/fimmu.2023.1240678] [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: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 09/05/2023] Open
Abstract
Background Therapeutic cancer vaccination against mutant calreticulin (CALR) in patients with CALR-mutant (CALRmut) myeloproliferative neoplasms (MPN) induces strong T-cell responses against mutant CALR yet fails to demonstrate clinical activity. Infiltration of tumor specific T cells into the tumor microenvironment is needed to attain a clinical response to therapeutic cancer vaccination. Aim Determine if CALRmut specific T cells isolated from vaccinated patients enrich in the bone marrow upon completion of vaccination and explore possible explanations for the lack of enrichment. Methods CALRmut specific T cells from four of ten vaccinated patients were expanded, enriched, and analyzed by T-cell receptor sequencing (TCRSeq). The TCRs identified were used as fingerprints of CALRmut specific T cells. Bone marrow aspirations from the four patients were acquired at baseline and at the end of trial. T cells were enriched from the bone marrow aspirations and analyzed by TCRSeq to identify the presence and fraction of CALRmut specific T cells at the two different time points. In silico calculations were performed to calculate the ratio between transformed cells and effector cells in patients with CALRmut MPN. Results The fraction of CALRmut specific T cells in the bone marrow did not increase upon completion of the vaccination trial. In general, the T cell repertoire in the bone marrow remains relatively constant through the vaccination trial. The enriched and expanded CALRmut specific T cells recognize peripheral blood autologous CALRmut cells. In silico analyses demonstrate a high imbalance in the fraction of CALRmut cells and CALRmut specific effector T-cells in peripheral blood. Conclusion CALRmut specific T cells do not enrich in the bone marrow after therapeutic cancer peptide vaccination against mutant CALR. The specific T cells recognize autologous peripheral blood derived CALRmut cells. In silico analyses demonstrate a high imbalance between the number of transformed cells and CALRmut specific effector T-cells in the periphery. We suggest that the high burden of transformed cells in the periphery compared to the number of effector cells could impact the ability of specific T cells to enrich in the bone marrow.
Collapse
Affiliation(s)
- Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Morten Andersen
- Centre for Mathematical Modeling – Human Health and Disease, IMFUFA, Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Sofie Traynor
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Shamaila Munir Ahmad
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Thomas Landkildehus Lisle
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Jacob Handlos Grauslund
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Johnny T. Ottesen
- Centre for Mathematical Modeling – Human Health and Disease, IMFUFA, Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Morten Frier Gjerstorff
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Soyfer EM, Fleischman AG. Myeloproliferative neoplasms - blurring the lines between cancer and chronic inflammatory disorder. Front Oncol 2023; 13:1208089. [PMID: 37361587 PMCID: PMC10288874 DOI: 10.3389/fonc.2023.1208089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Myeloproliferative Neoplasm (MPN) is a group of chronic blood cancers that arise from a hematopoietic stem cell (HSC) clone with somatic mutations causing constitutive activation of myeloid cytokine receptor signaling. In addition to elevated blood cell counts, MPN typically presents with increased inflammatory signaling and inflammation symptoms. Therefore, while being a clonally derived neoplasm, MPN has much in common with chronic non-cancerous inflammatory conditions, such as rheumatoid arthritis, lupus, and many more. MPN and chronic inflammatory disease (CID) share similar chronicity, symptoms, dependency on the immune system, environmental triggers, and treatments. Overall, we will highlight the similarities between an MPN and CID. We highlight that while MPN is classified as a cancer, its behavior is more aligned to that of a chronic inflammatory disease. We propose that MPN should inhabit a fluid/spectrum between auto-inflammatory disease and cancer.
Collapse
Affiliation(s)
- Eli M. Soyfer
- School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Angela G. Fleischman
- School of Medicine, University of California, Irvine, Irvine, CA, United States
- Division of Hematology/Oncology, University of California (UC) Irvine Health, Irvine, CA, United States
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
27
|
Ivanov D, Milosevic Feenstra JD, Sadovnik I, Herrmann H, Peter B, Willmann M, Greiner G, Slavnitsch K, Hadzijusufovic E, Rülicke T, Dahlhoff M, Hoermann G, Machherndl‐Spandl S, Eisenwort G, Fillitz M, Sliwa T, Krauth M, Bettelheim P, Sperr WR, Koller E, Pfeilstöcker M, Gisslinger H, Keil F, Kralovics R, Valent P. Phenotypic characterization of disease-initiating stem cells in JAK2- or CALR-mutated myeloproliferative neoplasms. Am J Hematol 2023; 98:770-783. [PMID: 36814396 PMCID: PMC10952374 DOI: 10.1002/ajh.26889] [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: 11/30/2022] [Revised: 02/07/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023]
Abstract
Myeloproliferative neoplasms (MPN) are characterized by uncontrolled expansion of myeloid cells, disease-related mutations in certain driver-genes including JAK2, CALR, and MPL, and a substantial risk to progress to secondary acute myeloid leukemia (sAML). Although behaving as stem cell neoplasms, little is known about disease-initiating stem cells in MPN. We established the phenotype of putative CD34+ /CD38- stem cells and CD34+ /CD38+ progenitor cells in MPN. A total of 111 patients with MPN suffering from polycythemia vera, essential thrombocythemia, or primary myelofibrosis (PMF) were examined. In almost all patients tested, CD34+ /CD38- stem cells expressed CD33, CD44, CD47, CD52, CD97, CD99, CD105, CD117, CD123, CD133, CD184, CD243, and CD274 (PD-L1). In patients with PMF, MPN stem cells often expressed CD25 and sometimes also CD26 in an aberrant manner. MPN stem cells did not exhibit substantial amounts of CD90, CD273 (PD-L2), CD279 (PD-1), CD366 (TIM-3), CD371 (CLL-1), or IL-1RAP. The phenotype of CD34+ /CD38- stem cells did not change profoundly during progression to sAML. The disease-initiating capacity of putative MPN stem cells was confirmed in NSGS mice. Whereas CD34+ /CD38- MPN cells engrafted in NSGS mice, no substantial engraftment was produced by CD34+ /CD38+ or CD34- cells. The JAK2-targeting drug fedratinib and the BRD4 degrader dBET6 induced apoptosis and suppressed proliferation in MPN stem cells. Together, MPN stem cells display a unique phenotype, including cytokine receptors, immune checkpoint molecules, and other clinically relevant target antigens. Phenotypic characterization of neoplastic stem cells in MPN and sAML should facilitate their enrichment and the development of stem cell-eradicating (curative) therapies.
Collapse
Affiliation(s)
- Daniel Ivanov
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | | | - Irina Sadovnik
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | - Harald Herrmann
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Department of Radiation OncologyMedical University of ViennaViennaAustria
| | - Barbara Peter
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | - Michael Willmann
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Department for Companion Animals, Clinical Unit for Internal MedicineUniversity of Veterinary Medicine ViennaViennaAustria
| | - Georg Greiner
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
- Ihr Labor, Medical Diagnostic LaboratoriesViennaAustria
| | - Katharina Slavnitsch
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Emir Hadzijusufovic
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Department for Companion Animals, Clinical Unit for Internal MedicineUniversity of Veterinary Medicine ViennaViennaAustria
| | - Thomas Rülicke
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Department of Biomedical SciencesUniversity of Veterinary Medicine ViennaViennaAustria
| | - Maik Dahlhoff
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Gregor Hoermann
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- MLL Munich Leukemia LaboratoryMunichGermany
| | - Sigrid Machherndl‐Spandl
- Hospital Ordensklinikum Elisabethinen LinzLinzAustria
- Johannes Kepler University, Medical FacultyLinzAustria
| | - Gregor Eisenwort
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Michael Fillitz
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Thamer Sliwa
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Maria‐Theresa Krauth
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | | | - Wolfgang R. Sperr
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | - Elisabeth Koller
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Michael Pfeilstöcker
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Heinz Gisslinger
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
| | - Felix Keil
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Robert Kralovics
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| |
Collapse
|
28
|
Ciarambino T, Crispino P, Minervini G, Giordano M. Cerebral Sinus Vein Thrombosis and Gender: A Not Entirely Casual Relationship. Biomedicines 2023; 11:1280. [PMID: 37238951 PMCID: PMC10216036 DOI: 10.3390/biomedicines11051280] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/13/2023] [Accepted: 04/22/2023] [Indexed: 05/28/2023] Open
Abstract
Cerebral sinus venous thrombosis (CSVT) is a relatively rare acute disorder of cerebral circulation, but it can potentially be associated with serious sequelae and a poor prognosis. The neurological manifestations associated with it are often not adequately taken into consideration given the extreme variability and nuances of its clinical presentation and given the need for radiological methods suitable for this type of diagnosis. CSVT is usually more common in women, but so far there are little data available in the literature on sex-specific characteristics regarding this pathology. CSVT is the result of multiple conditions and is therefore to be considered a multifactorial disease where at least one risk factor is present in over 80% of cases. From the literature, we learn that congenital or acquired prothrombotic states are to be considered extremely associated with the occurrence of an acute episode of CSVT and its recurrences. It is, therefore, necessary to fully know the origins and natural history of CSVT, in order to implement the diagnostic and therapeutic pathways of these neurological manifestations. In this report, we summarize the main causes of CSVT considering the possible influence of gender, bearing in mind that most of the causes listed above are pathological conditions closely linked to the female sex.
Collapse
Affiliation(s)
- Tiziana Ciarambino
- Internal Medicine Department, Hospital of Marcianise, ASL Caserta, 81024 Caserta, Italy
| | - Pietro Crispino
- Internal Medicine Department, Hospital of Latina, ASL Latina, 04100 Latina, Italy
| | - Giovanni Minervini
- Emergency Department, Hospital of Lagonegro, AOR San Carlo, 85042 Lagonegro, Italy
| | - Mauro Giordano
- Advanced Medical and Surgical Sciences Department, University of Campania, L. Vanvitelli, 81100 Naples, Italy
| |
Collapse
|
29
|
Yang FC, Agosto-Peña J. Epigenetic regulation by ASXL1 in myeloid malignancies. Int J Hematol 2023; 117:791-806. [PMID: 37062051 DOI: 10.1007/s12185-023-03586-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/02/2023] [Accepted: 03/22/2023] [Indexed: 04/17/2023]
Abstract
Myeloid malignancies are clonal hematopoietic disorders that are comprised of a spectrum of genetically heterogeneous disorders, including myelodysplastic syndromes (MDS), myeloproliferative neoplasms (MPN), chronic myelomonocytic leukemia (CMML), and acute myeloid leukemia (AML). Myeloid malignancies are characterized by excessive proliferation, abnormal self-renewal, and/or differentiation defects of hematopoietic stem cells (HSCs) and myeloid progenitor cells hematopoietic stem/progenitor cells (HSPCs). Myeloid malignancies can be caused by genetic and epigenetic alterations that provoke key cellular functions, such as self-renewal, proliferation, biased lineage commitment, and differentiation. Advances in next-generation sequencing led to the identification of multiple mutations in myeloid neoplasms, and many new gene mutations were identified as key factors in driving the pathogenesis of myeloid malignancies. The polycomb protein ASXL1 was identified to be frequently mutated in all forms of myeloid malignancies, with mutational frequencies of 20%, 43%, 10%, and 20% in MDS, CMML, MPN, and AML, respectively. Significantly, ASXL1 mutations are associated with a poor prognosis in all forms of myeloid malignancies. The fact that ASXL1 mutations are associated with poor prognosis in patients with CMML, MDS, and AML, points to the possibility that ASXL1 mutation is a key factor in the development of myeloid malignancies. This review summarizes the recent advances in understanding myeloid malignancies with a specific focus on ASXL1 mutations.
Collapse
Affiliation(s)
- Feng-Chun Yang
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
- Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| | - Joel Agosto-Peña
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| |
Collapse
|
30
|
Zhang R, Liu Y, Song H, Tan Y, Jin W, Zhao H, Liu X, Li J, Wang G, Chen L, Liang Y, Wang K. Clonal evolution analysis of a rare acute promyelocytic leukemia patient transforming from essential thrombocythemia. Ann Hematol 2023; 102:981-984. [PMID: 36749404 DOI: 10.1007/s00277-023-05112-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/28/2023] [Indexed: 02/08/2023]
Affiliation(s)
- Rong Zhang
- Department of Hematology, Xi'an Gaoxin Hospital, Xi'an, China
| | - Yabin Liu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd, Shanghai, 200025, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Huan Song
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd, Shanghai, 200025, China
| | - Yun Tan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd, Shanghai, 200025, China
| | - Wen Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd, Shanghai, 200025, China
| | - Huiying Zhao
- Department of Hematology, Xi'an Gaoxin Hospital, Xi'an, China
| | - Xiaowu Liu
- Department of Hematology, Xi'an Gaoxin Hospital, Xi'an, China
| | - Jing Li
- Department of Hematology, Xi'an Gaoxin Hospital, Xi'an, China
| | - Gangfeng Wang
- Department of Hematology, Hematology Hospital of Xi'an International Medical Center, Xi'an, China
| | - Li Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd, Shanghai, 200025, China
| | - Yingmin Liang
- Department of Hematology, Hematology Hospital of Xi'an International Medical Center, Xi'an, China.
| | - Kankan Wang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China. .,Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd, Shanghai, 200025, China.
| |
Collapse
|
31
|
Foßelteder J, Pabst G, Sconocchia T, Schlacher A, Auinger L, Kashofer K, Beham-Schmid C, Trajanoski S, Waskow C, Schöll W, Sill H, Zebisch A, Wölfler A, Thomas D, Reinisch A. Human gene-engineered calreticulin mutant stem cells recapitulate MPN hallmarks and identify targetable vulnerabilities. Leukemia 2023; 37:843-853. [PMID: 36813992 PMCID: PMC10079532 DOI: 10.1038/s41375-023-01848-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023]
Abstract
Calreticulin (CALR) mutations present the main oncogenic drivers in JAK2 wildtype (WT) myeloproliferative neoplasms (MPN), including essential thrombocythemia and myelofibrosis, where mutant (MUT) CALR is increasingly recognized as a suitable mutation-specific drug target. However, our current understanding of its mechanism-of-action is derived from mouse models or immortalized cell lines, where cross-species differences, ectopic over-expression and lack of disease penetrance are hampering translational research. Here, we describe the first human gene-engineered model of CALR MUT MPN using a CRISPR/Cas9 and adeno-associated viral vector-mediated knock-in strategy in primary human hematopoietic stem and progenitor cells (HSPCs) to establish a reproducible and trackable phenotype in vitro and in xenografted mice. Our humanized model recapitulates many disease hallmarks: thrombopoietin-independent megakaryopoiesis, myeloid-lineage skewing, splenomegaly, bone marrow fibrosis, and expansion of megakaryocyte-primed CD41+ progenitors. Strikingly, introduction of CALR mutations enforced early reprogramming of human HSPCs and the induction of an endoplasmic reticulum stress response. The observed compensatory upregulation of chaperones revealed novel mutation-specific vulnerabilities with preferential sensitivity of CALR mutant cells to inhibition of the BiP chaperone and the proteasome. Overall, our humanized model improves purely murine models and provides a readily usable basis for testing of novel therapeutic strategies in a human setting.
Collapse
Affiliation(s)
- Johannes Foßelteder
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Gabriel Pabst
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria.,Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria.,Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Tommaso Sconocchia
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Angelika Schlacher
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Lisa Auinger
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Karl Kashofer
- Diagnostic & Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Medical University of Graz, Graz, Austria
| | - Claudia Waskow
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany.,Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich-Schiller-University, Jena, Germany
| | - Wolfgang Schöll
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Heinz Sill
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Armin Zebisch
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria.,Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Albert Wölfler
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Daniel Thomas
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Andreas Reinisch
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria. .,Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria.
| |
Collapse
|
32
|
Chia YC, Siti Asmaa MJ, Ramli M, Woon PY, Johan MF, Hassan R, Islam MA. Molecular Genetics of Thrombotic Myeloproliferative Neoplasms: Implications in Precision Oncology. Diagnostics (Basel) 2023; 13:163. [PMID: 36611455 PMCID: PMC9818412 DOI: 10.3390/diagnostics13010163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 01/06/2023] Open
Abstract
Classical BCR-ABL-negative myeloproliferative neoplasms (MPN) include polycythaemia vera, essential thrombocythaemia, and primary myelofibrosis. Unlike monogenic disorders, a more complicated series of genetic mutations are believed to be responsible for MPN with various degrees of thromboembolic and bleeding complications. Thrombosis is one of the early manifestations in patients with MPN. To date, the driver genes responsible for MPN include JAK2, CALR, MPL, TET2, ASXL1, and MTHFR. Affords have been done to elucidate these mutations and the incidence of thromboembolic events. Several lines of evidence indicate that mutations in JAK2, MPL, TET2 and ASXL1 gene and polymorphisms in several clotting factors (GPIa, GPIIa, and GPIIIa) are associated with the occurrence and prevalence of thrombosis in MPN patients. Some polymorphisms within XRCC1, FBG, F2, F5, F7, F12, MMP9, HPA5, MTHFR, SDF-1, FAS, FASL, TERT, ACE, and TLR4 genes may also play a role in MPN manifestation. This review aims to provide an insightful overview on the genetic perspective of thrombotic complications in patients with MPN.
Collapse
Affiliation(s)
- Yuh Cai Chia
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Mat Jusoh Siti Asmaa
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Marini Ramli
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Peng Yeong Woon
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan
| | - Muhammad Farid Johan
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Rosline Hassan
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Md Asiful Islam
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
33
|
Qanneta R, Sopena E, Bové-Aleu E. [Erythromelalgia as a first clinical manifestation of polycythemia vera in an elderly patient]. Rev Esp Geriatr Gerontol 2023; 58:55-57. [PMID: 36379727 DOI: 10.1016/j.regg.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/15/2022]
Affiliation(s)
- Rami Qanneta
- Hospital Sociosanitari Francolí, Gestió i Prestació de Serveis de Salut (GiPSS), Tarragona, España.
| | - Eugenia Sopena
- Hospital Sociosanitari Francolí, Gestió i Prestació de Serveis de Salut (GiPSS), Tarragona, España
| | - Elisa Bové-Aleu
- Hospital Sociosanitari Francolí, Gestió i Prestació de Serveis de Salut (GiPSS), Tarragona, España
| |
Collapse
|
34
|
Wang Z, Wang Z, Zhang Z, Li J, Pan Z, Liu A, Lu J, Guo J, Zu M, Xu H. Establishment and validation of a prediction model for the first recurrence of Budd-Chiari syndrome after endovascular treatment: a large sample size, single-center retrospective study. Hepatol Int 2022; 17:159-169. [PMID: 36567373 PMCID: PMC9895038 DOI: 10.1007/s12072-022-10464-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/24/2022] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To investigate the independent risk factors for the first recurrence after endovascular management in patients with Budd-Chiari syndrome (BCS), and to establish a prediction model for predicting recurrence in target patients. METHODS BCS patients who underwent endovascular treatment in the Affiliated Hospital of Xuzhou Medical University from January 2010 to December 2015 were retrospectively examined, with their clinical, laboratory test, and imaging data collected and analyzed. Independent risk factors for recurrence were identified, and a prediction model was established and validated. RESULTS A total of 450 patients met the filtering criteria, and 102 recurred during the follow-up. The median follow-up time was 87 months, ranging from 1 to 137 months. The 1-, 3-, 5- and 10-year cumulative recurrence rate was 9.11% (6.41-11.73%), 17.35% (13.77-20.78%), 20.10% (16.30-23.72%), and 23.06% (18.86-27.04%), respectively. Liver cirrhosis, ascites, thrombosis, and all the main intrahepatic drainage veins obstructed (obstructed HV + AHV) are independent risk factors, while age is an independent protective factor. The prediction model was named MRBET. Based on the model, the risk score of each patient equals (-0.385981 * Age/10) + (0.0404184 * PT) + (0.0943423 * CRE/10) + (0.0157053 * LDH/10) + (0.592179 * LC) + (0.896034 * Ascites) + (0.691346 * Thrombosis) + (0.886741 * obstructed HV + AHV), and those in the high-risk group (risk score ≥ 1.57) were more likely to recur than those in the low-risk group (HR = 6.911, p < 0.001). The MRBET model is also available as a web tool at https://mrbet.shinyapps.io/dynnomapp . CONCLUSION Liver cirrhosis, ascites, thrombosis, and obstructed HV + AHV are independent risk factors for the first recurrence; age is an independent protective factor. The prediction model can effectively and conveniently predict the risk of recurrence and screen out patients at a high recurrence risk.
Collapse
Affiliation(s)
- Zhongkai Wang
- Department of Interventional Radiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006 Jiangsu China ,Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009 Jiangsu China
| | - Ziwei Wang
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433 China
| | - Zhiyuan Zhang
- Department of Interventional Oncology, The Second Affiliated Hospital of Xuzhou Medical University, 32 Meijian Road, Xuzhou, 221006 Jiangsu China
| | - Jiandong Li
- Department of Interventional Radiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006 Jiangsu China
| | - Zhiyang Pan
- Department of Interventional Radiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006 Jiangsu China
| | - Ang Liu
- Department of Interventional Radiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006 Jiangsu China ,Department of Structural Heart Disease, Cardiovascular Institute and Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing, 100037 China
| | - Jian Lu
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009 Jiangsu China
| | - Jinhe Guo
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009 Jiangsu China
| | - Maoheng Zu
- Department of Interventional Radiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006 Jiangsu China
| | - Hao Xu
- Department of Interventional Radiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006 Jiangsu China
| |
Collapse
|
35
|
Hsieh HH, Yao H, Ma Y, Zhang Y, Xiao X, Stephens H, Wajahat N, Chung SS, Xu L, Xu J, Rampal RK, Huang LJS. Epo-IGF1R cross talk expands stress-specific progenitors in regenerative erythropoiesis and myeloproliferative neoplasm. Blood 2022; 140:2371-2384. [PMID: 36054916 PMCID: PMC9837451 DOI: 10.1182/blood.2022016741] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/22/2022] [Indexed: 01/21/2023] Open
Abstract
We found that in regenerative erythropoiesis, the erythroid progenitor landscape is reshaped, and a previously undescribed progenitor population with colony-forming unit-erythroid (CFU-E) activity (stress CFU-E [sCFU-E]) is expanded markedly to restore the erythron. sCFU-E cells are targets of erythropoietin (Epo), and sCFU-E expansion requires signaling from the Epo receptor (EpoR) cytoplasmic tyrosines. Molecularly, Epo promotes sCFU-E expansion via JAK2- and STAT5-dependent expression of IRS2, thus engaging the progrowth signaling from the IGF1 receptor (IGF1R). Inhibition of IGF1R and IRS2 signaling impairs sCFU-E cell growth, whereas exogenous IRS2 expression rescues cell growth in sCFU-E expressing truncated EpoR-lacking cytoplasmic tyrosines. This sCFU-E pathway is the major pathway involved in erythrocytosis driven by the oncogenic JAK2 mutant JAK2(V617F) in myeloproliferative neoplasm. Inability to expand sCFU-E cells by truncated EpoR protects against JAK2(V617F)-driven erythrocytosis. In samples from patients with myeloproliferative neoplasm, the number of sCFU-E-like cells increases, and inhibition of IGR1R and IRS2 signaling blocks Epo-hypersensitive erythroid cell colony formation. In summary, we identified a new stress-specific erythroid progenitor cell population that links regenerative erythropoiesis to pathogenic erythrocytosis.
Collapse
Affiliation(s)
- Hsi-Hsien Hsieh
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | - Huiyu Yao
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | - Yue Ma
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | - Yuannyu Zhang
- Children’s Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX
| | - Xue Xiao
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX
| | - Helen Stephens
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Naureen Wajahat
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | - Stephen S. Chung
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Lin Xu
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX
| | - Jian Xu
- Children’s Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX
- Department of Pediatrics, Harold C. Simmons Comprehensive Cancer Center and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Raajit K. Rampal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | |
Collapse
|
36
|
Li XQ, Liu JJ, Liu CC. Case report: Double L611S/V617L JAK2 mutation in a patient with polycythemia vera originally diagnosed with essential thrombocythemia. Front Oncol 2022; 12:937362. [DOI: 10.3389/fonc.2022.937362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022] Open
Abstract
Double JAK2 mutations have rarely been described in myeloproliferative neoplasms (MPNs) and are demonstrated to be associated with the polycythemia vera (PV) phenotype. Here, we first report a case of a PV patient with a de novo double L611S/V617L in cis mutation of JAK2. A 40-year-old woman was admitted to the hospital with massive splenomegaly, multiple splenic infarcts, and abdominal pain. She had a 4-year history of erythrocytosis with an antecedent 10-year history of thrombocytosis before coming to our hospital. She was diagnosed with JAK2 L611S/V617L double-mutant PV after a detailed medical examination in 2017. According to the literature, IFNα therapy can induce clinical, hematological, histopathological, and occasionally molecular remission in individuals with MPNs. Our report demonstrates that combination therapy with ruxolitinib and IFNα can lead to a substantial reduction in JAK2 L611S/V617L double-mutant allele burden.
Collapse
|
37
|
Larsen MK, Skov V, Kjær L, Møller‐Palacino NA, Pedersen RK, Andersen M, Ottesen JT, Cordua S, Poulsen HE, Dahl M, Knudsen TA, Eickhardt‐Dalbøge CS, Koschmieder S, Pedersen KM, Çolak Y, Bojesen SE, Nordestgaard BG, Stiehl T, Hasselbalch HC, Ellervik C. Clonal haematopoiesis of indeterminate potential and impaired kidney function-A Danish general population study with 11 years follow-up. Eur J Haematol 2022; 109:576-585. [PMID: 36054308 PMCID: PMC9804367 DOI: 10.1111/ejh.13845] [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: 05/26/2022] [Accepted: 08/08/2022] [Indexed: 01/05/2023]
Abstract
The myeloproliferative neoplasms are associated with chronic kidney disease but whether clonal haematopoiesis of indeterminate potential (CHIP) is associated with impaired kidney function is unknown. In the Danish General Suburban Population Study (N = 19 958) from 2010 to 2013, 645 individuals were positive for JAK2V617F (N = 613) or CALR (N = 32) mutations. Mutation-positive individuals without haematological malignancy were defined as having CHIP (N = 629). We used multiple and inverse probability weighted (IPW)-adjusted linear regression analysis to estimate adjusted mean (95% confidence interval) differences in estimated glomerular filtration rate (eGFR; ml/min/1.73 m2 ) by mutation status, variant allele frequency (VAF%), blood cell counts, and neutrophil-to-lymphocyte ratio (NLR). We performed 11-year longitudinal follow-up of eGFR in all individuals. Compared to CHIP-negative individuals, the mean differences in eGFR were -5.6 (-10.3, -0.8, p = .02) for CALR, -11.9 (-21.4, -2.4, p = 0.01) for CALR type 2, and -10.1 (-18.1, -2.2, p = .01) for CALR with VAF ≥ 1%. The IPW-adjusted linear regression analyses showed similar results. NLR was negatively associated with eGFR. Individuals with CALR type 2 had a worse 11-year longitudinal follow-up on eGFR compared to CHIP-negative individuals (p = .004). In conclusion, individuals with CALR mutations, especially CALR type 2, had impaired kidney function compared to CHIP-negative individuals as measured by a lower eGFR at baseline and during 11-year follow-up.
Collapse
Affiliation(s)
- Morten K. Larsen
- Department of HaematologyZealand University HospitalRoskildeDenmark,Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Vibe Skov
- Department of HaematologyZealand University HospitalRoskildeDenmark
| | - Lasse Kjær
- Department of HaematologyZealand University HospitalRoskildeDenmark
| | | | | | - Morten Andersen
- Department of Science and EnvironmentRoskilde UniversityRoskildeDenmark
| | - Johnny T. Ottesen
- Department of Science and EnvironmentRoskilde UniversityRoskildeDenmark
| | - Sabrina Cordua
- Department of HaematologyCopenhagen University Hospital RigshospitaletCopenhagenDenmark
| | - Henrik E. Poulsen
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of EndocrinologyCopenhagen University Hospital, Bispebjerg Frederiksberg HospitalCopenhagenDenmark,Department of CardiologyCopenhagen University Hospital, Nordsjællands HospitalHillerødDenmark
| | - Morten Dahl
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Clinical BiochemistryZealand University HospitalKøgeDenmark
| | - Trine A. Knudsen
- Department of HaematologyZealand University HospitalRoskildeDenmark,Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Christina Schjellerup Eickhardt‐Dalbøge
- Department of HaematologyZealand University HospitalRoskildeDenmark,Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Steffen Koschmieder
- Department of Haematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of MedicineRWTH Aachen UniversityAachenGermany
| | - Kasper M. Pedersen
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Clinical Biochemistry and the Copenhagen General Population StudyCopenhagen University Hospital, Herlev and Gentofte HospitalHerlevDenmark
| | - Yunus Çolak
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Clinical Biochemistry and the Copenhagen General Population StudyCopenhagen University Hospital, Herlev and Gentofte HospitalHerlevDenmark,Department of Respiratory MedicineCopenhagen University Hospital, Herlev and Gentofte HospitalDenmark
| | - Stig E. Bojesen
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Clinical Biochemistry and the Copenhagen General Population StudyCopenhagen University Hospital, Herlev and Gentofte HospitalHerlevDenmark
| | - Børge G. Nordestgaard
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Clinical Biochemistry and the Copenhagen General Population StudyCopenhagen University Hospital, Herlev and Gentofte HospitalHerlevDenmark
| | - Thomas Stiehl
- Department of Science and EnvironmentRoskilde UniversityRoskildeDenmark,Institute for Computational Biomedicine ‐ Disease ModellingFaculty of Medicine, RWTH Aachen UniversityAachenGermany
| | - Hans C. Hasselbalch
- Department of HaematologyZealand University HospitalRoskildeDenmark,Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Christina Ellervik
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Data SupportRegion ZealandDenmark,Department of PathologyHarvard Medical SchoolBostonUSA
| |
Collapse
|
38
|
Van Avondt K, Strecker J, Tulotta C, Minnerup J, Schulz C, Soehnlein O. Neutrophils in aging and aging‐related pathologies. Immunol Rev 2022; 314:357-375. [PMID: 36315403 DOI: 10.1111/imr.13153] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Over the past millennia, life expectancy has drastically increased. While a mere 25 years during Bronze and Iron ages, life expectancy in many European countries and in Japan is currently above 80 years. Such an increase in life expectancy is a result of improved diet, life style, and medical care. Yet, increased life span and aging also represent the most important non-modifiable risk factors for several pathologies including cardiovascular disease, neurodegenerative diseases, and cancer. In recent years, neutrophils have been implicated in all of these pathologies. Hence, this review provides an overview of how aging impacts neutrophil production and function and conversely how neutrophils drive aging-associated pathologies. Finally, we provide a perspective on how processes of neutrophil-driven pathologies in the context of aging can be targeted therapeutically.
Collapse
Affiliation(s)
- Kristof Van Avondt
- Institute of Experimental Pathology (ExPat), Centre of Molecular Biology of Inflammation (ZMBE) University of Münster Münster Germany
| | - Jan‐Kolja Strecker
- Department of Neurology with Institute of Translational Neurology University Hospital Münster Münster Germany
| | - Claudia Tulotta
- Institute of Experimental Pathology (ExPat), Centre of Molecular Biology of Inflammation (ZMBE) University of Münster Münster Germany
| | - Jens Minnerup
- Department of Neurology with Institute of Translational Neurology University Hospital Münster Münster Germany
| | - Christian Schulz
- Department of Medicine I University Hospital, Ludwig Maximilian University Munich Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance Munich Germany
| | - Oliver Soehnlein
- Institute of Experimental Pathology (ExPat), Centre of Molecular Biology of Inflammation (ZMBE) University of Münster Münster Germany
- Department of Physiology and Pharmacology (FyFa) Karolinska Institute Stockholm Sweden
| |
Collapse
|
39
|
Htun HL, Lian W, Wong J, Tan EJ, Foo LL, Ong KH, Lim WY. Classic myeloproliferative neoplasms in Singapore: A population-based study on incidence, trends, and survival from 1968 to 2017. Cancer Epidemiol 2022; 79:102175. [DOI: 10.1016/j.canep.2022.102175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022]
|
40
|
Ibarra J, Elbanna YA, Kurylowicz K, Ciboddo M, Greenbaum HS, Arellano NS, Rodriguez D, Evers M, Bock-Hughes A, Liu C, Smith Q, Lutze J, Baumeister J, Kalmer M, Olschok K, Nicholson B, Silva D, Maxwell L, Dowgielewicz J, Rumi E, Pietra D, Casetti IC, Catricala S, Koschmieder S, Gurbuxani S, Schneider RK, Oakes SA, Elf SE. Type I but Not Type II Calreticulin Mutations Activate the IRE1α/XBP1 Pathway of the Unfolded Protein Response to Drive Myeloproliferative Neoplasms. Blood Cancer Discov 2022; 3:298-315. [PMID: 35405004 PMCID: PMC9338758 DOI: 10.1158/2643-3230.bcd-21-0144] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/21/2022] [Accepted: 04/08/2022] [Indexed: 01/09/2023] Open
Abstract
Approximately 20% of patients with myeloproliferative neoplasms (MPN) harbor mutations in the gene calreticulin (CALR), with 80% of those mutations classified as either type I or type II. While type II CALR-mutant proteins retain many of the Ca2+ binding sites present in the wild-type protein, type I CALR-mutant proteins lose these residues. The functional consequences of this differential loss of Ca2+ binding sites remain unexplored. Here, we show that the loss of Ca2+ binding residues in the type I mutant CALR protein directly impairs its Ca2+ binding ability, which in turn leads to depleted endoplasmic reticulum (ER) Ca2+ and subsequent activation of the IRE1α/XBP1 pathway of the unfolded protein response. Genetic or pharmacologic inhibition of IRE1α/XBP1 signaling induces cell death in type I mutant but not type II mutant or wild-type CALR-expressing cells, and abrogates type I mutant CALR-driven MPN disease progression in vivo. SIGNIFICANCE Current targeted therapies for CALR-mutated MPNs are not curative and fail to differentiate between type I- versus type II-driven disease. To improve treatment strategies, it is critical to identify CALR mutation type-specific vulnerabilities. Here we show that IRE1α/XBP1 represents a unique, targetable dependency specific to type I CALR-mutated MPNs. This article is highlighted in the In This Issue feature, p. 265.
Collapse
Affiliation(s)
- Juan Ibarra
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Yassmin A. Elbanna
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Katarzyna Kurylowicz
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Michele Ciboddo
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Harrison S. Greenbaum
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Nicole S. Arellano
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Deborah Rodriguez
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Maria Evers
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Althea Bock-Hughes
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Chenyu Liu
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Quinn Smith
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Julian Lutze
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois
| | - 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
| | - Milena Kalmer
- 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
| | - Kathrin Olschok
- 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
| | - Benjamin Nicholson
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Diane Silva
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Luke Maxwell
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Jonathan Dowgielewicz
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Elisa Rumi
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Daniela Pietra
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | | | - Silvia Catricala
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - 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
| | | | - Rebekka K. Schneider
- Department of Cell Biology, Institute for Biomedical Technologies, RWTH Aachen University, Aachen, Germany
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Scott A. Oakes
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Shannon E. Elf
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| |
Collapse
|
41
|
Interferon-alpha2 treatment of patients with polycythemia vera and related neoplasms favorably impacts deregulation of oxidative stress genes and antioxidative defense mechanisms. PLoS One 2022; 17:e0270669. [PMID: 35771847 PMCID: PMC9246201 DOI: 10.1371/journal.pone.0270669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/14/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic inflammation is considered a major driving force for clonal expansion and evolution in the Philadelphia-negative myeloproliferative neoplasms, which include essential thrombocythemia, polycythemia vera and primary myelofibrosis (MPNs). One of the key mutation drivers is the JAK2V617F mutation, which has been shown to induce the generation of reactive oxygen species (ROS). Using whole blood gene expression profiling, deregulation of several oxidative stress and anti-oxidative defense genes has been identified in MPNs, including significant downregulation of TP53, the NFE2L2 or NRF2 genes. These genes have a major role for maintaining genomic stability, regulation of the oxidative stress response and in modulating migration or retention of hematopoietic stem cells. Therefore, their deregulation might give rise to increasing genomic instability, increased chronic inflammation and disease progression with egress of hematopoietic stem cells from the bone marrow to seed in the spleen, liver and elsewhere. Interferon-alpha2 (rIFNα) is increasingly being recognized as the drug of choice for the treatment of patients with MPNs. Herein, we report the first gene expression profiling study on the impact of rIFNα upon oxidative stress and antioxidative defense genes in patients with MPNs (n = 33), showing that rIFNα downregulates several upregulated oxidative stress genes and upregulates downregulated antioxidative defense genes. Treatment with rIFNα induced upregulation of 19 genes in ET and 29 genes in PV including CXCR4 and TP53. In conclusion, this rIFNα- mediated dampening of genotoxic damage to hematopoietic cells may ultimately diminish the risk of additional mutations and accordingly clonal evolution and disease progression towards myelofibrotic and leukemic transformation.
Collapse
|
42
|
Gigoux M, Holmström MO, Zappasodi R, Park JJ, Pourpe S, Bozkus CC, Mangarin LMB, Redmond D, Verma S, Schad S, George MM, Venkatesh D, Ghosh A, Hoyos D, Molvi Z, Kamaz B, Marneth AE, Duke W, Leventhal MJ, Jan M, Ho VT, Hobbs GS, Knudsen TA, Skov V, Kjær L, Larsen TS, Hansen DL, Lindsley RC, Hasselbalch H, Grauslund JH, Lisle TL, Met Ö, Wilkinson P, Greenbaum B, Sepulveda MA, Chan T, Rampal R, Andersen MH, Abdel-Wahab O, Bhardwaj N, Wolchok JD, Mullally A, Merghoub T. Calreticulin mutant myeloproliferative neoplasms induce MHC-I skewing, which can be overcome by an optimized peptide cancer vaccine. Sci Transl Med 2022; 14:eaba4380. [PMID: 35704596 PMCID: PMC11182673 DOI: 10.1126/scitranslmed.aba4380] [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: 11/02/2022]
Abstract
The majority of JAK2V617F-negative myeloproliferative neoplasms (MPNs) have disease-initiating frameshift mutations in calreticulin (CALR), resulting in a common carboxyl-terminal mutant fragment (CALRMUT), representing an attractive source of neoantigens for cancer vaccines. However, studies have shown that CALRMUT-specific T cells are rare in patients with CALRMUT MPN for unknown reasons. We examined class I major histocompatibility complex (MHC-I) allele frequencies in patients with CALRMUT MPN from two independent cohorts. We observed that MHC-I alleles that present CALRMUT neoepitopes with high affinity are underrepresented in patients with CALRMUT MPN. We speculated that this was due to an increased chance of immune-mediated tumor rejection by individuals expressing one of these MHC-I alleles such that the disease never clinically manifested. As a consequence of this MHC-I allele restriction, we reasoned that patients with CALRMUT MPN would not efficiently respond to a CALRMUT fragment cancer vaccine but would when immunized with a modified CALRMUT heteroclitic peptide vaccine approach. We found that heteroclitic CALRMUT peptides specifically designed for the MHC-I alleles of patients with CALRMUT MPN efficiently elicited a CALRMUT cross-reactive CD8+ T cell response in human peripheral blood samples but not to the matched weakly immunogenic CALRMUT native peptides. We corroborated this effect in vivo in mice and observed that C57BL/6J mice can mount a CD8+ T cell response to the CALRMUT fragment upon immunization with a CALRMUT heteroclitic, but not native, peptide. Together, our data emphasize the therapeutic potential of heteroclitic peptide-based cancer vaccines in patients with CALRMUT MPN.
Collapse
Affiliation(s)
- Mathieu Gigoux
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Morten O. Holmström
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Joseph J. Park
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Stephane Pourpe
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Levi M. B. Mangarin
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David Redmond
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Svena Verma
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Sara Schad
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Mariam M. George
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Divya Venkatesh
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Arnab Ghosh
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David Hoyos
- Computational Oncology, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zaki Molvi
- Weill Cornell Medicine, New York, NY 10065, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Baransel Kamaz
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anna E. Marneth
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - William Duke
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Max Jan
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Vincent T. Ho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Gabriela S. Hobbs
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Trine Alma Knudsen
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | | | - Dennis Lund Hansen
- Department of Hematology, Odense University Hospital, Odense 5000, Denmark
| | - R. Coleman Lindsley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Hans Hasselbalch
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Jacob H. Grauslund
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Thomas L. Lisle
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Özcan Met
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Patrick Wilkinson
- Janssen Oncology Therapeutic Area, Janssen Research and Development, LLC, Pharmaceutical Companies of Johnson & Johnson, Spring House, PA 19002, USA
| | - Benjamin Greenbaum
- Computational Oncology, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medicine, Physiology, Biophysics and Systems Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Manuel A. Sepulveda
- Janssen Oncology Therapeutic Area, Janssen Research and Development, LLC, Pharmaceutical Companies of Johnson & Johnson, Spring House, PA 19002, USA
| | - Timothy Chan
- Weill Cornell Medical College, New York, NY 10065, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Raajit Rampal
- Human Oncology and Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mads H. Andersen
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nina Bhardwaj
- Parker Institute for Cancer Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jedd D. Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Ann Mullally
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute, Cambridge, MA 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
43
|
Henry SP, Liosi ME, Ippolito JA, Cutrona KJ, Krimmer SG, Newton AS, Schlessinger J, Jorgensen WL. Conversion of a False Virtual Screen Hit into Selective JAK2 JH2 Domain Binders Using Convergent Design Strategies. ACS Med Chem Lett 2022; 13:819-826. [PMID: 35586418 PMCID: PMC9109162 DOI: 10.1021/acsmedchemlett.2c00051] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
The Janus kinase 2 (JAK2) pseudokinase domain (JH2) is an ATP-binding domain that regulates the activity of the catalytic tyrosine kinase domain (JH1). Dysregulation of JAK2 JH1 signaling caused by the V617F mutation in JH2 is implicated in various myeloproliferative neoplasms. To explore if JAK2 activity can be modulated by a small molecule binding to the ATP site in JH2, we have developed several ligand series aimed at selectively targeting the JAK2 JH2 domain. We report here the evolution of a false virtual screen hit into a new JAK2 JH2 series. Optimization guided by computational modeling has yielded analogues with nanomolar affinity for the JAK2 JH2 domain and >100-fold selectivity for the JH2 domain over the JH1 domain. A crystal structure for one of the potent compounds bound to JAK2 JH2 clarifies the origins of the strong binding and selectivity. The compounds expand the platform for seeking molecules to regulate JAK2 signaling, including V617F JAK2 hyperactivation.
Collapse
Affiliation(s)
- Sean P. Henry
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Maria-Elena Liosi
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Joseph A. Ippolito
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Kara J. Cutrona
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Stefan G. Krimmer
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
- Department
of Pharmacology, Yale University School
of Medicine, New Haven, Connecticut 06520-8066, United States
| | - Ana S. Newton
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Joseph Schlessinger
- Department
of Pharmacology, Yale University School
of Medicine, New Haven, Connecticut 06520-8066, United States
| | - William L. Jorgensen
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| |
Collapse
|
44
|
Wang X, Rampal RK, Hu CS, Tripodi J, Farnoud N, Petersen B, Rossi MR, Patel M, McGovern E, Najfeld V, Iancu-Rubin C, Lu M, Davis A, Kremyanskaya M, Weinberg RS, Mascarenhas J, Hoffman R. Characterization of disease-propagating stem cells responsible for myeloproliferative neoplasm-blast phase. JCI Insight 2022; 7:e156534. [PMID: 35259128 PMCID: PMC9089790 DOI: 10.1172/jci.insight.156534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/02/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic myeloproliferative neoplasms (MPN) frequently evolve to a blast phase (BP) that is almost uniformly resistant to induction chemotherapy or hypomethylating agents. We explored the functional properties, genomic architecture, and cell of origin of MPN-BP initiating cells (IC) using a serial NSG mouse xenograft transplantation model. Transplantation of peripheral blood mononuclear cells (MNC) from 7 of 18 patients resulted in a high degree of leukemic cell chimerism and recreated clinical characteristics of human MPN-BP. The function of MPN-BP ICs was not dependent on the presence of JAK2V617F, a driver mutation associated with the initial underlying MPN. By contrast, multiple MPN-BP IC subclones coexisted within MPN-BP MNCs characterized by different myeloid malignancy gene mutations and cytogenetic abnormalities. MPN-BP ICs in 4 patients exhibited extensive proliferative and self-renewal capacity, as demonstrated by their ability to recapitulate human MPN-BP in serial recipients. These MPN-BP IC subclones underwent extensive continuous clonal competition within individual xenografts and across multiple generations, and their subclonal dynamics were consistent with functional evolution of MPN-BP IC. Finally, we show that MPN-BP ICs originate from not only phenotypically identified hematopoietic stem cells, but also lymphoid-myeloid progenitor cells, which were each characterized by differences in MPN-BP initiating activity and self-renewal capacity.
Collapse
Affiliation(s)
- Xiaoli Wang
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| | - Raajit K. Rampal
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Cing Siang Hu
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| | - Joseph Tripodi
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| | - Noushin Farnoud
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Bruce Petersen
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| | - Michael R. Rossi
- Genetics and Genomic Sciences, ISMMS, New York, New York
- Sema4, Stamford, Connecticut, USA
| | - Minal Patel
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Erin McGovern
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Vesna Najfeld
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| | - Camelia Iancu-Rubin
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| | - Min Lu
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| | - Andrew Davis
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| | - Marina Kremyanskaya
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| | | | - John Mascarenhas
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| | - Ronald Hoffman
- Division of Hematology/Medical Oncology/Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York, USA
| |
Collapse
|
45
|
Molecular Pathogenesis of Myeloproliferative Neoplasms: From Molecular Landscape to Therapeutic Implications. Int J Mol Sci 2022; 23:ijms23094573. [PMID: 35562964 PMCID: PMC9100530 DOI: 10.3390/ijms23094573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 12/27/2022] Open
Abstract
Despite distinct clinical entities, the myeloproliferative neoplasms (MPN) share morphological similarities, propensity to thrombotic events and leukemic evolution, and a complex molecular pathogenesis. Well-known driver mutations, JAK2, MPL and CALR, determining constitutive activation of JAK-STAT signaling pathway are the hallmark of MPN pathogenesis. Recent data in MPN patients identified the presence of co-occurrence somatic mutations associated with epigenetic regulation, messenger RNA splicing, transcriptional mechanism, signal transduction, and DNA repair mechanism. The integration of genetic information within clinical setting is already improving patient management in terms of disease monitoring and prognostic information on disease progression. Even the current therapeutic approaches are limited in disease-modifying activity, the expanding insight into the genetic basis of MPN poses novel candidates for targeted therapeutic approaches. This review aims to explore the molecular landscape of MPN, providing a comprehensive overview of the role of drive mutations and additional mutations, their impact on pathogenesis as well as their prognostic value, and how they may have future implications in therapeutic management.
Collapse
|
46
|
The Use of Allogeneic Hematopoietic Stem Cell Transplantation in Primary Myelofibrosis. J Pers Med 2022; 12:jpm12040571. [PMID: 35455686 PMCID: PMC9025208 DOI: 10.3390/jpm12040571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/04/2022] [Accepted: 03/14/2022] [Indexed: 01/27/2023] Open
Abstract
Primary myelofibrosis (PMF) is a BCR-ABL1 negative myeloproliferative neoplasm characterized by clonal proliferation of myeloid cells. This leads to reactive bone marrow fibrosis, ultimately resulting in progressive marrow failure, hepatosplenomegaly, and extramedullary hematopoiesis. PMF is considered the most aggressive of the BCR-ABL1 negative myeloproliferative neoplasms with the least favorable prognosis. Constitutional symptoms are common, which can impact an individual’s quality of life and leukemic transformation remains an important cause of death in PMF patients. The development of the Janus kinase 2 (JAK2) inhibitors have provided a good option for management of PMF-related symptoms. Unfortunately, these agents have not been shown to improve overall survival or significantly alter the course of disease. Allogenic hematopoietic stem cell transplantation (allo-HSCT) remains the only curative treatment option in PMF. However, allo-HSCT is associated with significant treatment-related morbidity and mortality and has historically been reserved for younger, high-risk patients. This review examines patient, disease, and transplant-specific factors which may impact transplant-related outcomes in PMF. Through the vast improvements in donor selection, conditioning regimens, and post-transplant care, allo-HSCT may provide a safe and effective curative option for a broader range of PMF patients in the future.
Collapse
|
47
|
Torres DG, Paes J, da Costa AG, Malheiro A, Silva GV, Mourão LPDS, Tarragô AM. JAK2 Variant Signaling: Genetic, Hematologic and Immune Implication in Chronic Myeloproliferative Neoplasms. Biomolecules 2022; 12:291. [PMID: 35204792 PMCID: PMC8961666 DOI: 10.3390/biom12020291] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
The JAK2V617F variant constitutes a genetic alteration of higher frequency in BCR/ABL1 negative chronic myeloproliferative neoplasms, which is caused by a substitution of a G ˃ T at position 1849 and results in the substitution of valine with phenylalanine at codon 617 of the polypeptide chain. Clinical, morphological and molecular genetic features define the diagnosis criteria of polycythemia vera, essential thrombocythemia and primary myelofibrosis. Currently, JAK2V617F is associated with clonal hematopoiesis, genomic instability, dysregulations in hemostasis and immune response. JAK2V617F clones induce an inflammatory immune response and lead to a process of immunothrombosis. Recent research has shown great interest in trying to understand the mechanisms associated with JAK2V617F signaling and activation of cellular and molecular responses that progressively contribute to the development of inflammatory and vascular conditions in association with chronic myeloproliferative neoplasms. Thus, the aim of this review is to describe the main genetic, hematological and immunological findings that are linked to JAK2 variant signaling in chronic myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Dania G. Torres
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
| | - Jhemerson Paes
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
| | - Allyson G. da Costa
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| | - Adriana Malheiro
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| | - George V. Silva
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Fundação Oswaldo Cruz–Instituto Leônidas e Maria Deane (Fiocruz), Manaus 69027-070, AM, Brazil
- Fundação Centro de Controle de Oncologia do Amazonas (FCECON), Manaus 69040-010, AM, Brazil
| | - Lucivana P. de Souza Mourão
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
| | - Andréa M. Tarragô
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (FHEMOAM), Manaus 69050-001, AM, Brazil
| |
Collapse
|
48
|
Celik H, Krug E, Zhang CR, Han W, Issa N, Koh WK, Bjeije H, Kukhar O, Allen M, Li T, Fisher DAC, Fowles JS, Wong TN, Stubbs MC, Koblish HK, Oh ST, Challen GA. A Humanized Animal Model Predicts Clonal Evolution and Therapeutic Vulnerabilities in Myeloproliferative Neoplasms. Cancer Discov 2021; 11:3126-3141. [PMID: 34193440 PMCID: PMC8716669 DOI: 10.1158/2159-8290.cd-20-1652] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 06/04/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022]
Abstract
Myeloproliferative neoplasms (MPN) are chronic blood diseases with significant morbidity and mortality. Although sequencing studies have elucidated the genetic mutations that drive these diseases, MPNs remain largely incurable with a significant proportion of patients progressing to rapidly fatal secondary acute myeloid leukemia (sAML). Therapeutic discovery has been hampered by the inability of genetically engineered mouse models to generate key human pathologies such as bone marrow fibrosis. To circumvent these limitations, here we present a humanized animal model of myelofibrosis (MF) patient-derived xenografts (PDX). These PDXs robustly engrafted patient cells that recapitulated the patient's genetic hierarchy and pathologies such as reticulin fibrosis and propagation of MPN-initiating stem cells. The model can select for engraftment of rare leukemic subclones to identify patients with MF at risk for sAML transformation and can be used as a platform for genetic target validation and therapeutic discovery. We present a novel but generalizable model to study human MPN biology. SIGNIFICANCE Although the genetic events driving MPNs are well defined, therapeutic discovery has been hampered by the inability of murine models to replicate key patient pathologies. Here, we present a PDX system to model human myelofibrosis that reproduces human pathologies and is amenable to genetic and pharmacologic manipulation. This article is highlighted in the In This Issue feature, p. 2945.
Collapse
Affiliation(s)
- Hamza Celik
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ethan Krug
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Christine R Zhang
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Wentao Han
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Nancy Issa
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Won Kyun Koh
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Hassan Bjeije
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ostap Kukhar
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Maggie Allen
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Tiandao Li
- Center of Regenerative Medicine, Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| | - Daniel A C Fisher
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jared S Fowles
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Terrence N Wong
- Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | | | | | - Stephen T Oh
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Grant A Challen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
49
|
Bohiltea RE, Niculescu-Mizil E, Mihai BM, Furtunescu F, Ducu I, Munteanu O, Georgescu TA, Grigoriu C. Polycythemia vera in pregnancy represents a challenge for a multidisciplinary collaboration: A case report and literature review. Exp Ther Med 2021; 23:19. [PMID: 34815771 DOI: 10.3892/etm.2021.10941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/24/2021] [Indexed: 01/10/2023] Open
Abstract
Polycythemia vera (PV) is a rare chronic myeloproliferative neoplasm which represents an additional thrombotic factor in pregnancy. PV may be difficult to diagnose, particularly as its incidence is extremely uncommon among young women. The main diagnostic method involves a bone marrow biopsy, and high hemoglobin and platelet counts are usually indicative of the condition, after excluding other more frequent pathologies. PV is associated with a high risk of thrombosis, particularly in pregnancy, and requires anti-platelet treatment. At present, only a limited number of PV cases in pregnancy have been reported in the literature, at least to the best of our knowledge, with the largest case series being a retrospective study that included 25 pregnancies in 15 women. The present study describes the case of a patient diagnosed with JAK2-positive PV and also discusses this rare condition with particular focus on the following: i) The management of PV in pregnancy along with the additional pathologies in this specific case; and ii) the particularities of the pregnancy course. By identifying women suffering from PV superimposed by other possible procoagulant factors and applying the latest standard in healthcare, fetal and maternal prognosis may be significantly improved.
Collapse
Affiliation(s)
- Roxana Elena Bohiltea
- Department of Obstetrics and Gynecology, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania.,Department of Obstetrics and Gynecology, Life Memorial Hospital, 012244 Bucharest, Romania
| | | | - Bianca Margareta Mihai
- Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania
| | - Florentina Furtunescu
- Department of Public Health and Management, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050463 Bucharest, Romania
| | - Ionita Ducu
- Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania
| | - Octavian Munteanu
- Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania.,Department of Anatomy, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Tiberiu Augustin Georgescu
- Department of Pathology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050463 Bucharest, Romania
| | - Corina Grigoriu
- Department of Obstetrics and Gynecology, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania
| |
Collapse
|
50
|
Clonal hematopoiesis with JAK2V617F promotes pulmonary hypertension with ALK1 upregulation in lung neutrophils. Nat Commun 2021; 12:6177. [PMID: 34702814 PMCID: PMC8548396 DOI: 10.1038/s41467-021-26435-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/05/2021] [Indexed: 12/17/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive cardiopulmonary disease characterized by pulmonary arterial remodeling. Clonal somatic mutations including JAK2V617F, the most frequent driver mutation among myeloproliferative neoplasms, have recently been identified in healthy individuals without hematological disorders. Here, we reveal that clonal hematopoiesis with JAK2V617F exacerbates PH and pulmonary arterial remodeling in mice. JAK2V617F-expressing neutrophils specifically accumulate in pulmonary arterial regions, accompanied by increases in neutrophil-derived elastase activity and chemokines in chronic hypoxia-exposed JAK2V617F transgenic (JAK2V617F) mice, as well as recipient mice transplanted with JAK2V617F bone marrow cells. JAK2V617F progressively upregulates Acvrl1 (encoding ALK1) during the differentiation from bone marrow stem/progenitor cells peripherally into mature neutrophils of pulmonary arterial regions. JAK2V617F-mediated STAT3 phosphorylation upregulates ALK1-Smad1/5/8 signaling. ALK1/2 inhibition completely prevents the development of PH in JAK2V617F mice. Finally, our prospective clinical study identified JAK2V617F-positive clonal hematopoiesis is more common in PH patients than in healthy subjects. These findings indicate that clonal hematopoiesis with JAK2V617F causally leads to PH development associated with ALK1 upregulation.
Collapse
|