1
|
Harrison A, Zhang T, Batra G. A case of hemichorea-hemiballismus secondary to a subacute hemorrhage in a patient with essential thrombocythemia: a case report. J Med Case Rep 2025; 19:142. [PMID: 40148955 PMCID: PMC11948710 DOI: 10.1186/s13256-025-05161-x] [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: 08/06/2024] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Hemiballismus is a rare movement disorder characterized by violent, involuntary movements, usually resulting from a stroke affecting the subthalamic nucleus. Risperidone, an atypical antipsychotic, has shown effectiveness in managing these symptoms. CASE PRESENTATION We present the case of a 75-year-old Asian female patient of Cambodian origin with hemichorea-hemiballismus secondary to a subacute hemorrhage in the left basal ganglia, in the context of essential thrombocythemia. The patient responded remarkably well to risperidone. The patient's story, along with video documentation of her condition and response to treatment, is discussed. CONCLUSION This case highlights the potential of risperidone in treating hemichorea-hemiballismus, providing a nonsurgical option for managing this debilitating condition.
Collapse
Affiliation(s)
- Anil Harrison
- Department of Internal Medicine, Midwestern University, Glendale, AZ, 85308, USA.
- Department of Internal Medicine, University of Central Florida/HCA, Pensacola, FL, 32514, USA.
| | - Tiantian Zhang
- Department of Internal Medicine, University of Central Florida/HCA, Pensacola, FL, 32514, USA
| | - Gary Batra
- Department of Internal Medicine, University of Central Florida/HCA, Pensacola, FL, 32514, USA
| |
Collapse
|
2
|
Carturan A, Morè S, Poloni A, Rupoli S, Morsia E. Shaping the Future of Myeloproliferative Neoplasm Therapy: Immune-Based Strategies and Targeted Innovations. Cancers (Basel) 2024; 16:4113. [PMID: 39682299 DOI: 10.3390/cancers16234113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Numerous cutting-edge immunotherapy approaches have been developed for hematological malignancies, such as immune-checkpoint inhibitors for lymphomas, chimeric antigen receptor (CAR)-T-cell treatments for B-cell cancers, and monoclonal antibody therapies for acute myeloid leukemia (AML). However, achieving similar breakthroughs in MPNs has proven challenging. The key obstacles include the absence of universally expressed and MPN-specific surface markers, significant cellular and molecular variability among both individual patients and across different MPN subtypes, and the failure of treatments to stimulate an anti-tumor immune response due to the immune system disruptions caused by the myeloid neoplasm. Currently, there are several innovative therapies in clinical trials for MPNs. These include new JAK inhibitors with greater specificity for JAK2, as well as "add-on" medications designed to enhance the effectiveness of ruxolitinib, in both patients who are new to the drug and in those who have shown suboptimal responses. Additionally, there is ongoing exploration of novel therapeutic targets. In this review, we will explore the immunotherapy approaches that are currently used in clinical practice for MPNs, as well as emerging strategies that are likely to change the treatment of these diseases in the coming years.
Collapse
Affiliation(s)
- Alberto Carturan
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Hematology and Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sonia Morè
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, 60126 Ancona, Italy
- Hematology Clinic, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Antonella Poloni
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, 60126 Ancona, Italy
- Hematology Clinic, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Serena Rupoli
- Hematology Clinic, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Erika Morsia
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, 60126 Ancona, Italy
- Hematology Clinic, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| |
Collapse
|
3
|
Ganesan S, Awan-Toor S, Guidez F, Maslah N, Rahimy R, Aoun C, Gou P, Guiguen C, Soret J, Ravdan O, Bisio V, Dulphy N, Lobry C, Schlageter MH, Souyri M, Giraudier S, Kiladjian JJ, Chomienne C, Cassinat B. Comprehensive analysis of mesenchymal cells reveals a dysregulated TGF-β/WNT/HOXB7 axis in patients with myelofibrosis. JCI Insight 2024; 9:e173665. [PMID: 39470742 PMCID: PMC11623938 DOI: 10.1172/jci.insight.173665] [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: 07/12/2023] [Accepted: 10/22/2024] [Indexed: 11/01/2024] Open
Abstract
Despite the advances in the understanding and treatment of myeloproliferative neoplasm (MPN), the disease remains incurable with the risk of evolution to acute myeloid leukemia or myelofibrosis (MF). Unfortunately, the evolution of the disease to MF remains poorly understood, impeding preventive and therapeutic options. Recent studies in solid tumor microenvironment and organ fibrosis have shed instrumental insights on their respective pathogenesis and drug resistance, yet such precise data are lacking in MPN. In this study, through a patient sample-driven transcriptomic and epigenetic description of the MF microenvironment landscape and cell-based analyses, we identify homeobox B7 (HOXB7) overexpression and more precisely a potentially novel TGF-β/WNT/HOXB7 pathway as associated to a pro-fibrotic and pro-osteoblastic biased differentiation of mesenchymal stromal cells (MSCs). Using gene-based and chemical inhibition of this pathway, we reversed the abnormal phenotype of MSCs from patients with MF, providing the MPN field a potentially novel target to prevent and manage evolution to MF.
Collapse
Affiliation(s)
- Saravanan Ganesan
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Sarah Awan-Toor
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Fabien Guidez
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- INSERM U1232/LNC, Team Epi2THM, Université Bourgogne Franche-Comté, Dijon, France
| | - Nabih Maslah
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- Service de Biologie Cellulaire, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Rifkath Rahimy
- Laboratoire de recherche en génétique et hématologie translationnelle, Institut Gonçalo Moniz, Salvador, Bahia, Brazil
| | - Céline Aoun
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Panhong Gou
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Chloé Guiguen
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Juliette Soret
- INSERM CIC 1427, Université Paris Cité, Centre d’Investigations Cliniques, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Odonchimeg Ravdan
- Service de Biologie Cellulaire, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Valeria Bisio
- INSERM UMRS 1160, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris, France
| | - Nicolas Dulphy
- INSERM UMRS 1160, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris, France
- Laboratoire d’Immunologie et d’Histocompatibilite, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Camille Lobry
- INSERM U944, CNRS UMR7212, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris, France
| | | | - Michèle Souyri
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Stéphane Giraudier
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- Service de Biologie Cellulaire, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Jean-Jacques Kiladjian
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- INSERM CIC 1427, Université Paris Cité, Centre d’Investigations Cliniques, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Christine Chomienne
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Bruno Cassinat
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- Service de Biologie Cellulaire, Hôpital Saint-Louis, AP-HP, Paris, France
| |
Collapse
|
4
|
Krecak I, Lekovic D, Bogdanovic A, Lucijanic M. High red cell mass and high plasma volume are independently associated with thrombotic risk in polycythemia vera. Leuk Lymphoma 2024; 65:1724-1728. [PMID: 38899619 DOI: 10.1080/10428194.2024.2368639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Affiliation(s)
- Ivan Krecak
- Department of Internal Medicine, General Hospital of Sibenik-Knin County, Sibenik, Croatia
- Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- University of Applied Sciences, Sibenik, Croatia
| | - Danijela Lekovic
- Clinic of Hematology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Andrija Bogdanovic
- Clinic of Hematology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marko Lucijanic
- Division of Hematology, University Hospital Dubrava, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
5
|
Leiva O, Liu O, Zhou S, How J, Lee M, Hobbs G. Myeloproliferative Neoplasms and Cardiovascular Disease: A Review. Curr Treat Options Oncol 2024; 25:1257-1267. [PMID: 39278999 DOI: 10.1007/s11864-024-01255-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2024] [Indexed: 09/18/2024]
Abstract
OPINION STATEMENT Myeloproliferative neoplasms (MPN) are a heterogenous group of disorders of clonal hematopoiesis characterized by constitutive activation of the JAK/STAT signaling pathway leading to proliferation of blood cells. Cardiovascular disease (CVD) contributes significantly to the morbidity and mortality of patients with MPN. Particularly well-known CVD complications of MPNs are arterial and venous thrombotic events. However, MPNs are also associated with other forms of CVD including atrial fibrillation, heart failure, and pulmonary hypertension. Recent studies have characterized outcomes of patients with MPN and CVD, including acute myocardial infarction (AMI), heart failure, atrial fibrillation, and pulmonary hypertension. Additionally, optimal cardiovascular disease prevention strategies in patients with MPN are not yet clear. Further investigation is warranted to improve CVD outcomes in patients with MPN. Clinicians should be aware of cardiovascular complications of MPN, including thrombotic as well as non-thrombotic complications (heart failure, arrhythmias, pulmonary hypertension).
Collapse
Affiliation(s)
- Orly Leiva
- Department of Medicine, Division of Cardiology, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Medicine, Section of Cardiology - Heart Failure, University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA.
| | - Olivia Liu
- Department of Medicine, Division of Cardiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sophia Zhou
- Department of Medicine, Division of Cardiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Joan How
- Department of Medicine, Division of Hematology and Oncology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michelle Lee
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gabriela Hobbs
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Ümit EG, Baysal M, Kırkızlar HO, Demir AM. Myeloproliferative Neoplasm Symptom Assessment Total Symptom Score (MPN-SAF TSS) in Chronic Myeloproliferative Neoplasms with Relation to Genetic Burden and Thrombosis. Turk J Haematol 2024; 41:175-181. [PMID: 38801033 PMCID: PMC11589363 DOI: 10.4274/tjh.galenos.2024.2024.0011] [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: 01/07/2024] [Accepted: 05/25/2024] [Indexed: 05/29/2024] Open
Abstract
The Myeloproliferative Neoplasm Symptom Assessment Total Symptom Score (MPN-SAF TSS) is a surrogate marker for symptom evaluation in chronic myeloproliferative neoplasms (MPNs). However, insufficient data are available regarding the relationship among the MPN-SAF TSS, JAK2 mutation allele burden, and thrombosis. In this retrospective analysis, we aimed to determine the genetic burdens, clinical features, and relationships with MPN-SAF TSS in MPN patients. One hundred thirty JAK2V617F-positive patients with MPNs were included in our study. We calculated the MPN-SAF TSS for all patients and compared it with their clinical characteristics. Patients with higher JAK2V617F mutation allele burden had higher MPN-SAF TSS values (p=0.008). Patients with thrombosis had higher MPN-SAF TSS than patients without thrombosis (p=0.003). The mean MPN-SAF TSS was higher in patients with primary myelofibrosis compared to those with polycythemia vera and essential thrombocythemia. Thrombosis was associated with increased symptom severity in several domains, including fatigue, abdominal discomfort, inactivity, night sweats, pruritus, weight loss, and early satiety. Additionally, an increase in JAK2 allele burden was observed with higher symptom scores. The MPN-SAF TSS proved to be a reliable tool for assessing symptom burden in Turkish MPN patients. Furthermore, the significant association between thrombosis occurrence and symptom severity suggests that thrombotic events may contribute to symptom development. Notably, increasing JAK2 allele burden was correlated with more severe symptoms, highlighting its potential role in predicting disease burden. This study emphasizes the importance of symptom assessment in MPN patients and supports the incorporation of the MPN-SAF TSS in routine clinical practice to enhance patient care and management.
Collapse
Affiliation(s)
- Elif Gülsüm Ümit
- Trakya University Faculty of Medicine, Department of Internal Medicine, Division of Hematology, Edirne, Türkiye
| | - Mehmet Baysal
- Ali Osman Sönmez Oncology Hospital, Clinic of Hematology, Bursa, Türkiye
| | - Hakkı Onur Kırkızlar
- Trakya University Faculty of Medicine, Department of Internal Medicine, Division of Hematology, Edirne, Türkiye
| | - Ahmet Muzaffer Demir
- Trakya University Faculty of Medicine, Department of Internal Medicine, Division of Hematology, Edirne, Türkiye
| |
Collapse
|
7
|
Kelliher S, Gamba S, Weiss L, Shen Z, Marchetti M, Schieppati F, Scaife C, Madden S, Bennett K, Fortune A, Maung S, Fay M, Ní Áinle F, Maguire P, Falanga A, Kevane B, Krishnan A. Platelet proteomic profiling reveals potential mediators of immunothrombosis and proteostasis in myeloproliferative neoplasms. Blood Adv 2024; 8:4276-4280. [PMID: 38861353 PMCID: PMC11372589 DOI: 10.1182/bloodadvances.2023012016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 05/17/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024] Open
Affiliation(s)
- Sarah Kelliher
- School of Medicine, University College Dublin, Dublin, Ireland
- Stanford University School of Medicine, Stanford University, Stanford, CA
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
| | - Sara Gamba
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Luisa Weiss
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Zhu Shen
- Stanford University School of Medicine, Stanford University, Stanford, CA
| | - Marina Marchetti
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Francesca Schieppati
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Caitriona Scaife
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kathleen Bennett
- School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Anne Fortune
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Su Maung
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Michael Fay
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Fionnuala Ní Áinle
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- Department of Haematology, Rotunda Hospital, Dublin, Ireland
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Patricia Maguire
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Institute for Discovery, University College Dublin, Dublin, Ireland
| | - Anna Falanga
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Barry Kevane
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
| | - Anandi Krishnan
- Stanford University School of Medicine, Stanford University, Stanford, CA
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ
- Stanford Cancer Institute, Stanford, CA
| |
Collapse
|
8
|
Tabata S, Yamashita Y, Inai Y, Morita S, Kosako H, Takagi T, Shide K, Manabe S, Matsuoka TA, Shimoda K, Sonoki T, Ihara Y, Tamura S. C-Mannosyl tryptophan is a novel biomarker for thrombocytosis of myeloproliferative neoplasms. Sci Rep 2024; 14:18858. [PMID: 39143127 PMCID: PMC11324734 DOI: 10.1038/s41598-024-69496-z] [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: 05/21/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024] Open
Abstract
C-Mannosyl tryptophan (CMW), a unique glycosylated amino acid, is considered to be produced by degradation of C-mannosylated proteins in living organism. Although protein C-mannosylation is involved in the folding and secretion of substrate proteins, the pathophysiological function in the hematological system is still unclear. This study aimed to assess CMW in the human hematological disorders. The serum CMW levels of 94 healthy Japanese workers were quantified using hydrophilic interaction liquid chromatography. Platelet count was positively correlated with serum CMW levels. The clinical significance of CMW in thrombocytosis of myeloproliferative neoplasms (T-MPN) including essential thrombocythemia (ET) were investigated. The serum CMW levels of the 34 patients with T-MPN who presented with thrombocytosis were significantly higher than those of the 52 patients with control who had other hematological disorders. In patients with T-MPN, serum CMW levels were inversely correlated with anemia, which was related to myelofibrosis (MF). Bone marrow biopsy samples were obtained from 18 patients with ET, and serum CMW levels were simultaneously measured. Twelve patients with bone marrow fibrosis had significantly higher CMW levels than 6 patients without bone marrow fibrosis. Collectively, these results suggested that CMW could be a novel biomarker to predict MF progression in T-MPN.
Collapse
Affiliation(s)
- Shotaro Tabata
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Yusuke Yamashita
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Yoko Inai
- Department of Biochemistry, Wakayama Medical University, Wakayama, Japan
| | - Shuhei Morita
- The First Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Hideki Kosako
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Tomoyuki Takagi
- The First Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
- Wakayama City Medical Association Seijinbyo Center, Wakayama, Japan
| | - Kotaro Shide
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Shino Manabe
- School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Science & Faculty of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Taka-Aki Matsuoka
- The First Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Takashi Sonoki
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Yoshito Ihara
- Department of Biochemistry, Wakayama Medical University, Wakayama, Japan.
| | - Shinobu Tamura
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan.
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
9
|
Todor SB, Ichim C, Boicean A, Mihaila RG. Cardiovascular Risk in Philadelphia-Negative Myeloproliferative Neoplasms: Mechanisms and Implications-A Narrative Review. Curr Issues Mol Biol 2024; 46:8407-8423. [PMID: 39194713 DOI: 10.3390/cimb46080496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs), encompassing disorders like polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are characterized by clonal hematopoiesis without the Philadelphia chromosome. The JAK2 V617F mutation is prevalent in PV, ET, and PMF, while mutations in MPL and CALR also play significant roles. These conditions predispose patients to thrombotic events, with PMF exhibiting the lowest survival among MPNs. Chronic inflammation, driven by cytokine release from aberrant leukocytes and platelets, amplifies cardiovascular risk through various mechanisms, including atherosclerosis and vascular remodeling. Additionally, MPN-related complications like pulmonary hypertension and cardiac fibrosis contribute to cardiovascular morbidity and mortality. This review consolidates recent research on MPNs' cardiovascular implications, emphasizing thrombotic risk, chronic inflammation, and vascular stiffness. Understanding these associations is crucial for developing targeted therapies and improving outcomes in MPN patients.
Collapse
Affiliation(s)
- Samuel Bogdan Todor
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Adrian Boicean
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | | |
Collapse
|
10
|
Cuenca-Zamora EJ, Guijarro-Carrillo PJ, López-Poveda MJ, Morales ML, Lozano ML, Gonzalez-Conejero R, Martínez C, Teruel-Montoya R, Ferrer-Marín F. miR-146a -/- mice model reveals that NF-κB inhibition reverts inflammation-driven myelofibrosis-like phenotype. Am J Hematol 2024; 99:1326-1337. [PMID: 38646919 DOI: 10.1002/ajh.27322] [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: 08/18/2023] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 04/23/2024]
Abstract
Emerging evidence shows the crucial role of inflammation (particularly NF-κB pathway) in the development and progression of myelofibrosis (MF), becoming a promising therapeutic target. Furthermore, tailoring treatment with currently available JAK inhibitors (such as ruxolitinib or fedratinib) does not modify the natural history of the disease and has important limitations, including cytopenias. Since recent studies have highlighted the role of miR-146a, a negative regulator of the NF-κB pathway, in the pathogenesis of MF; here we used miR-146a-/- (KO) mice, a MF-like model lacking driver mutations, to investigate whether pharmacological inhibition of JAK/STAT and/or NF-κB pathways may reverse the myelofibrotic phenotype of these mice. Specifically, we tested the JAK1/2 inhibitor, ruxolitinib; the NF-κB inhibitor via IKKα/β, BMS-345541; both inhibitors in combination; or a dual inhibitor of both pathways (JAK2/IRAK1), pacritinib. Although all treatments decreased spleen size and partially recovered its architecture, only NF-κB inhibition, either using BMS-345541 (alone or in combination) or pacritinib, resulted in a reduction of extramedullary hematopoiesis, bone marrow (BM) fibrosis and osteosclerosis, along with an attenuation of the exacerbated inflammatory state (via IL-1β and TNFα). However, although dual inhibitor improved anemia and reversed thrombocytopenia, the combined therapy worsened anemia by inducing BM hypoplasia. Both therapeutic options reduced NF-κB and JAK/STAT signaling in a context of JAK2V617F-driven clonal hematopoiesis. Additionally, combined treatment reduced both COL1A1 and IL-6 production in an in vitro model mimicking JAK2-driven fibrosis. In conclusion, NF-κB inhibition reduces, in vitro and in vivo, disease burden and BM fibrosis, which could provide benefits in myelofibrosis patients.
Collapse
Affiliation(s)
- Ernesto José Cuenca-Zamora
- Hematology Department, Hospital Universitario Morales-Meseguer, Centro Regional de Hemodonación, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
- CIBERER-ISCIII CB15/00055 (U765), Murcia, Spain
- Universidad de Murcia, Murcia, Spain
| | - Pedro J Guijarro-Carrillo
- Hematology Department, Hospital Universitario Morales-Meseguer, Centro Regional de Hemodonación, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
| | | | - María Luz Morales
- Hematology Department, Hospital Universitario Morales-Meseguer, Centro Regional de Hemodonación, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
| | - María Luisa Lozano
- Hematology Department, Hospital Universitario Morales-Meseguer, Centro Regional de Hemodonación, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
- CIBERER-ISCIII CB15/00055 (U765), Murcia, Spain
- Universidad de Murcia, Murcia, Spain
| | - Rocío Gonzalez-Conejero
- Hematology Department, Hospital Universitario Morales-Meseguer, Centro Regional de Hemodonación, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
- Universidad de Murcia, Murcia, Spain
| | - Constantino Martínez
- Hematology Department, Hospital Universitario Morales-Meseguer, Centro Regional de Hemodonación, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
| | - Raúl Teruel-Montoya
- Hematology Department, Hospital Universitario Morales-Meseguer, Centro Regional de Hemodonación, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
- CIBERER-ISCIII CB15/00055 (U765), Murcia, Spain
- Universidad de Murcia, Murcia, Spain
| | - Francisca Ferrer-Marín
- Hematology Department, Hospital Universitario Morales-Meseguer, Centro Regional de Hemodonación, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
- CIBERER-ISCIII CB15/00055 (U765), Murcia, Spain
- Universidad de Murcia, Murcia, Spain
- Universidad Católica San Antonio (UCAM), Murcia, Spain
| |
Collapse
|
11
|
Shahriyary F, Amirzargar MR, Vafajoo M, Kooshari A, Basi A, Razavi SM, Gharegozlou B, Shahidi M. Resveratrol and tetrahydroisoquinoline effects on neutrophil sensitivity to NETosis formation in low-risk essential thrombocythemia patients. Eur J Haematol 2024; 113:44-53. [PMID: 38544388 DOI: 10.1111/ejh.14196] [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: 12/03/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 06/04/2024]
Abstract
INTRODUCTION Recent studies scrutinize how NETosis (a unique cell death mechanism of neutrophil), impacts thrombosis patients with essential thrombocythemia (ET). This research evaluates the susceptibility of ET neutrophils to form NETs and tests two potential inhibitors, resveratrol (RSV) and tetrahydroisoquinoline (THIQ), in vitro. METHODS Platelet-rich plasma from low-risk ET patients was used, along with neutrophils from both patients and controls. NET formation assays, with or without RSV and THIQ treatment after LPS stimulation, were conducted in a CO2 incubator. Evaluation included flow cytometry and fluorescence microscopy for NET formation and ELISA for TNFα, IL8, and vWF:Ag levels in patient and control plasma. RESULTS Neutrophils from ET patients released more NETs than controls, confirmed by flow cytometry and fluorescence microscopy. Additionally, patients had significantly higher plasma levels of IL8 and TNFα compared to controls, while RSV was more effective than THIQ in reducing NETosis rates in these patients. CONCLUSIONS In ET patients, a platelet counts over 1 million indicates the need for preventive treatment against thrombotic events. Similarly, in this study, RSV and THIQ significantly reduced the rate of NETosis in ET patients with higher platelet counts, and this role was more prominent in the case of the second inhibitor (RSV).
Collapse
Affiliation(s)
- Fahimeh Shahriyary
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Amirzargar
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Vafajoo
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Kooshari
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Basi
- Department of Oncology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohsen Razavi
- Department of Oncology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Behnaz Gharegozlou
- Department of Immunology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Minoo Shahidi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Belmonte M, Cabrera-Cosme L, Øbro NF, Li J, Grinfeld J, Milek J, Bennett E, Irvine M, Shepherd MS, Cull AH, Boyd G, Riedel LM, Chi Che JL, Oedekoven CA, Baxter EJ, Green AR, Barlow JL, Kent DG. Increased CXCL10 (IP-10) is associated with advanced myeloproliferative neoplasms and its loss dampens erythrocytosis in mouse models. Exp Hematol 2024; 135:104246. [PMID: 38763471 DOI: 10.1016/j.exphem.2024.104246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 05/04/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Key studies in pre-leukemic disorders have linked increases in pro-inflammatory cytokines with accelerated phases of the disease, but the precise role of the cellular microenvironment in disease initiation and evolution remains poorly understood. In myeloproliferative neoplasms (MPNs), higher levels of specific cytokines have been previously correlated with increased disease severity (tumor necrosis factor-alpha [TNF-α], interferon gamma-induced protein-10 [IP-10 or CXCL10]) and decreased survival (interleukin 8 [IL-8]). Whereas TNF-α and IL-8 have been studied by numerous groups, there is a relative paucity of studies on IP-10 (CXCL10). Here we explore the relationship of IP-10 levels with detailed genomic and clinical data and undertake a complementary cytokine screen alongside functional assays in a wide range of MPN mouse models. Similar to patients, levels of IP-10 were increased in mice with more severe disease phenotypes (e.g., JAK2V617F/V617F TET2-/- double-mutant mice) compared with those with less severe phenotypes (e.g., CALRdel52 or JAK2+/V617F mice) and wild-type (WT) littermate controls. Although exposure to IP-10 did not directly alter proliferation or survival in single hematopoietic stem cells (HSCs) in vitro, IP-10-/- mice transplanted with disease-initiating HSCs developed an MPN phenotype more slowly, suggesting that the effect of IP-10 loss was noncell-autonomous. To explore the broader effects of IP-10 loss, we crossed IP-10-/- mice into a series of MPN mouse models and showed that its loss reduces the erythrocytosis observed in mice with the most severe phenotype. Together, these data point to a potential role for blocking IP-10 activity in the management of MPNs.
Collapse
Affiliation(s)
- Miriam Belmonte
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Lilia Cabrera-Cosme
- Department of Biology, Centre for Blood Research, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Nina F Øbro
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Juan Li
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Jacob Grinfeld
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, United Kingdom
| | - Joanna Milek
- Department of Biology, Centre for Blood Research, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Ellie Bennett
- Department of Biology, Centre for Blood Research, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Melissa Irvine
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Mairi S Shepherd
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Alyssa H Cull
- Department of Biology, Centre for Blood Research, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Grace Boyd
- Department of Biology, Centre for Blood Research, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Lisa M Riedel
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - James Lok Chi Che
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom; Department of Biology, Centre for Blood Research, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Caroline A Oedekoven
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - E Joanna Baxter
- Department of Haematology, Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, United Kingdom
| | - Anthony R Green
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, United Kingdom
| | - Jillian L Barlow
- Department of Biology, Centre for Blood Research, York Biomedical Research Institute, University of York, York, United Kingdom
| | - David G Kent
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom; Department of Biology, Centre for Blood Research, York Biomedical Research Institute, University of York, York, United Kingdom.
| |
Collapse
|
13
|
Xiong H, Zhang H, Bai J, Li Y, Li L, Zhang L. Associations of the circulating levels of cytokines with the risk of myeloproliferative neoplasms: a bidirectional mendelian-randomization study. BMC Cancer 2024; 24:531. [PMID: 38671390 PMCID: PMC11046808 DOI: 10.1186/s12885-024-12301-x] [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: 08/17/2023] [Accepted: 04/22/2024] [Indexed: 04/28/2024] Open
Abstract
OBJECTIVE In the pathogenesis of myeloproliferative neoplasms (MPN), inflammation plays an important role. However, it is unclear whether there is a causal link between inflammation and MPNs. We used a bidirectional, two-sample Mendelian randomization (MR) approach to investigate the causal relationship between systemic inflammatory cytokines and myeloproliferative neoplasms. METHODS A genome-wide association study (GWAS) of 8293 European participants identified genetic instrumental variables for circulating cytokines and growth factors. Summary statistics of MPN were obtained from a GWAS including 1086 cases and 407,155 controls of European ancestry. The inverse-variance-weighted method was mainly used to compute odds ratios (OR) and 95% confidence intervals (Cl). RESULTS Our results showed that higher Interleukin-2 receptor, alpha subunit (IL-2rα) levels, and higher Interferon gamma-induced protein 10 (IP-10) levels were associated with an increased risk of MPN (OR = 1.36,95%CI = 1.03-1.81, P = 0.032; OR = 1.55,95%CI = 1.09-2.22, P = 0.015; respectively).In addition, Genetically predicted MPN promotes expression of the inflammatory cytokines interleukin-10 (IL-10) (BETA = 0.033, 95% CI = 0.003 ~ 0.064, P = 0.032) and monokine induced by interferon-gamma (MIG) (BETA = 0.052, 95% CI = 0.002-0.102, P = 0.043) and, on activation, normal T cells express and secrete RANTES (BETA = 0.055, 95% CI = 0.0090.1, P = 0.018). CONCLUSION Our findings suggest that cytokines are essential to the pathophysiology of MPN. More research is required if these biomarkers can be used to prevent and treat MPN.
Collapse
Affiliation(s)
- Hao Xiong
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huitao Zhang
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jun Bai
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Yanhong Li
- 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
|
Zhang P, You N, Ding Y, Zhu W, Wang N, Xie Y, Huang W, Ren Q, Qin T, Fu R, Zhang L, Xiao Z, Cheng T, Ma X. Gadd45g insufficiency drives the pathogenesis of myeloproliferative neoplasms. Nat Commun 2024; 15:2989. [PMID: 38582902 PMCID: PMC10998908 DOI: 10.1038/s41467-024-47297-2] [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: 05/30/2023] [Accepted: 03/22/2024] [Indexed: 04/08/2024] Open
Abstract
Despite the identification of driver mutations leading to the initiation of myeloproliferative neoplasms (MPNs), the molecular pathogenesis of MPNs remains incompletely understood. Here, we demonstrate that growth arrest and DNA damage inducible gamma (GADD45g) is expressed at significantly lower levels in patients with MPNs, and JAK2V617F mutation and histone deacetylation contribute to its reduced expression. Downregulation of GADD45g plays a tumor-promoting role in human MPN cells. Gadd45g insufficiency in the murine hematopoietic system alone leads to significantly enhanced growth and self-renewal capacity of myeloid-biased hematopoietic stem cells, and the development of phenotypes resembling MPNs. Mechanistically, the pathogenic role of GADD45g insufficiency is mediated through a cascade of activations of RAC2, PAK1 and PI3K-AKT signaling pathways. These data characterize GADD45g deficiency as a novel pathogenic factor in MPNs.
Collapse
Affiliation(s)
- Peiwen Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Na You
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Yiyi Ding
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Wenqi Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Nan Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Yueqiao Xie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Wanling Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Tiejun Qin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Rongfeng Fu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.
| | - Zhijian Xiao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.
- Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China.
| | - Xiaotong Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China.
| |
Collapse
|
15
|
Rotta G, Gilardoni E, Ravazza D, Mock J, Seehusen F, Elsayed A, Puca E, De Luca R, Pellegrino C, Look T, Weiss T, Manz MG, Halin C, Neri D, Dakhel Plaza S. A novel strategy to generate immunocytokines with activity-on-demand using small molecule inhibitors. EMBO Mol Med 2024; 16:904-926. [PMID: 38448543 PMCID: PMC11018789 DOI: 10.1038/s44321-024-00034-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/08/2024] Open
Abstract
Cytokine-based therapeutics have been shown to mediate objective responses in certain tumor entities but suffer from insufficient selectivity, causing limiting toxicity which prevents dose escalation to therapeutically active regimens. The antibody-based delivery of cytokines significantly increases the therapeutic index of the corresponding payload but still suffers from side effects associated with peak concentrations of the product in blood upon intravenous administration. Here we devise a general strategy (named "Intra-Cork") to mask systemic cytokine activity without impacting anti-cancer efficacy. Our technology features the use of antibody-cytokine fusions, capable of selective localization at the neoplastic site, in combination with pathway-selective inhibitors of the cytokine signaling, which rapidly clear from the body. This strategy, exemplified with a tumor-targeted IL12 in combination with a JAK2 inhibitor, allowed to abrogate cytokine-driven toxicity without affecting therapeutic activity in a preclinical model of cancer. This approach is readily applicable in clinical practice.
Collapse
Affiliation(s)
- Giulia Rotta
- Philochem AG, CH-8112, Otelfingen, Switzerland
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | | | | | | | - Frauke Seehusen
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Abdullah Elsayed
- Philochem AG, CH-8112, Otelfingen, Switzerland
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093, Zurich, Switzerland
| | - Emanuele Puca
- Philochem AG, CH-8112, Otelfingen, Switzerland
- Philogen S.p.A, 53100, Siena, Italy
| | | | - Christian Pellegrino
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, CH-8091, Zurich, Switzerland
| | - Thomas Look
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, CH-8091, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, CH-8091, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, CH-8091, Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093, Zurich, Switzerland
| | - Dario Neri
- Philochem AG, CH-8112, Otelfingen, Switzerland.
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093, Zurich, Switzerland.
- Philogen S.p.A, 53100, Siena, Italy.
| | | |
Collapse
|
16
|
Kelliher S, Gamba S, Weiss L, Shen Z, Marchetti M, Schieppati F, Scaife C, Madden S, Bennett K, Fortune A, Maung S, Fay M, Ní Áinle F, Maguire P, Falanga A, Kevane B, Krishnan A. Platelet proteo-transcriptomic profiling validates mediators of thrombosis and proteostasis in patients with myeloproliferative neoplasms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563619. [PMID: 37961700 PMCID: PMC10634751 DOI: 10.1101/2023.10.23.563619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Patients with chronic Myeloproliferative Neoplasms (MPN) including polycythemia vera (PV) and essential thrombocythemia (ET) exhibit unique clinical features, such as a tendency toward thrombosis and hemorrhage, and risk of disease progression to secondary bone marrow fibrosis and/or acute leukemia. Although an increase in blood cell lineage counts (quantitative features) contribute to these morbid sequelae, the significant qualitative abnormalities of myeloid cells that contribute to vascular risk are not well understood. Here, we address this critical knowledge gap via a comprehensive and untargeted profiling of the platelet proteome in a large (n= 140) cohort of patients (from two independent sites) with an established diagnosis of PV and ET (and complement prior work on the MPN platelet transcriptome from a third site). We discover distinct MPN platelet protein expression and confirm key molecular impairments associated with proteostasis and thrombosis mechanisms of potential relevance to MPN pathology. Specifically, we validate expression of high-priority candidate markers from the platelet transcriptome at the platelet proteome (e.g., calreticulin (CALR), Fc gamma receptor (FcγRIIA) and galectin-1 (LGALS1) pointing to their likely significance in the proinflammatory, prothrombotic and profibrotic phenotypes in patients with MPN. Together, our proteo-transcriptomic study identifies the peripherally-derived platelet molecular profile as a potential window into MPN pathophysiology and demonstrates the value of integrative multi-omic approaches in gaining a better understanding of the complex molecular dynamics of disease.
Collapse
Affiliation(s)
- Sarah Kelliher
- School of Medicine, University College Dublin, Dublin, Ireland
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
| | - Sara Gamba
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Luisa Weiss
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Zhu Shen
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Marina Marchetti
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Francesca Schieppati
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Caitriona Scaife
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kathleen Bennett
- School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Anne Fortune
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Su Maung
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Michael Fay
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Fionnuala Ní Áinle
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Medicine, Royal College of Surgeons in Ireland
| | - Patricia Maguire
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Institute for Discovery, University College Dublin, Dublin, Ireland
| | - Anna Falanga
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
- University of Milano-Bicocca, Department of Medicine and Surgery, Monza, Italy
| | - Barry Kevane
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- UCD Conway SPHERE Research Group, University College Dublin, Dublin, Ireland
| | - Anandi Krishnan
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Rutgers University, Piscataway, NJ
- Stanford Cancer Institute, Stanford, CA, USA
| |
Collapse
|
17
|
Holik H, Krečak I, Lucijanić M, Samardžić I, Pilipac D, Vučinić Ljubičić I, Coha B, Kitter Pipić A, Miškić B, Zupančić-Šalek S. Hip and Knee Osteoarthritis in Patients with Chronic Myeloproliferative Neoplasms: A Cross-Sectional Study. Life (Basel) 2023; 13:1388. [PMID: 37374170 PMCID: PMC10300951 DOI: 10.3390/life13061388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a progressive degenerative disease with an inflammatory background. Chronic myeloproliferative neoplasms (MPN) are clonal hematopoietic disorders characterized by chronic inflammation and a tendency for connective tissue remodeling. AIM This study aimed to investigate the prevalence and associated risk factors of symptomatic OA (sOA) in MPN patients. PATIENTS AND METHODS A total of 100 consecutive MPN (39 essential-thrombocythemia, 34 polycythemia-vera, 27 myelofibrosis) patients treated in two community hematologic centers were cross-sectionally evaluated. Patients were required to have both symptoms attributable to hip and/or knee OA and radiographic confirmation to be considered as having sOA. RESULTS The prevalence of hip and/or knee sOA was significantly higher among MPN patients than the previously reported prevalence in the general population of similar age (61% vs. 22%, p < 0.001). Hip sOA was present in 50%, knee sOA in 51% and sOA of both localizations in 41% of patients. A high proportion of MPN patients had radiographic signs of hip OA (94%) and knee OA (98%) in the presence of attributable symptoms. Among the other factors, sOA was univariately associated with the presence of JAK2 mutation, myelofibrosis phenotype, older age, higher body weight, and higher MPN-SAF score (p < 0.050 for all analyses). In the multivariate analysis, older age (odds ratio = 1.19, 95% confidence interval-CI 1.06-1.33) and higher body weight (OR = 1.15, 95% CI 1.06-1.25) were recognized as independent risk factors for sOA. On the other hand, cytoreductive treatment was a protective factor for sOA (OR = 0.07, 95% CI 0.006-0.86). CONCLUSIONS The prevalence of sOA in MPN patients was higher than that in the general population and seems to correlate with older age, increased myeloproliferation and a higher inflammatory state. Whether cytoreductive treatment may postpone OA development in MPN patients warrants additional confirmation.
Collapse
Affiliation(s)
- Hrvoje Holik
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Ivan Krečak
- Department of Internal Medicine, General Hospital of Šibenik-Knin County, 22000 Šibenik, Croatia
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Marko Lucijanić
- University Hospital Dubrava, 10000 Zagreb, Croatia
- Faculty of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ivan Samardžić
- Department of Orthopaedic Surgery, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
| | - Danijel Pilipac
- Department of Orthopaedic Surgery, General Hospital of Šibenik-Knin County, 22000 Šibenik, Croatia
| | - Ivana Vučinić Ljubičić
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
| | - Božena Coha
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
| | - Alma Kitter Pipić
- Department of Laboratory Diagnostics, General Hospital ‘Dr Josip Benčević’, 35000 Slavonski Brod, Croatia
| | - Blaženka Miškić
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Silva Zupančić-Šalek
- Department of Hematology and Coagulation, University Hospital Holy Spirit, 10000 Zagreb, Croatia
| |
Collapse
|
18
|
Ross DM, Liang HPH, Iqra Z, Whittaker S, Tan CW, Dale BJ, Chen VM. Platelets from patients with myeloproliferative neoplasms have increased numbers of mitochondria that are hypersensitive to depolarization by thrombin. Sci Rep 2023; 13:9172. [PMID: 37280424 DOI: 10.1038/s41598-023-36266-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/31/2023] [Indexed: 06/08/2023] Open
Abstract
Thrombosis is one of the cardinal manifestations of myeloproliferative neoplasms (MPN). The mechanisms leading to a prothrombotic state in MPN are complex and remain poorly understood. Platelet mitochondria play a role in platelet activation, but their number and function have not been extensively explored in MPN to date. We observed an increased number of mitochondria in platelets from MPN patients compared with healthy donors. MPN patients had an increased proportion of dysfunctional platelet mitochondria. The fraction of platelets with depolarized mitochondria at rest was increased in essential thrombocythemia (ET) patients and the mitochondria were hypersensitive to depolarization following thrombin agonist stimulation. Live microscopy showed a stochastic process in which a higher proportion of individual ET platelets underwent mitochondrial depolarization and after a shorter agonist exposure compared to healthy donors. Depolarization was immediately followed by ballooning of the platelet membrane, which is a feature of procoagulant platelets. We also noted that the mitochondria of MPN patients were on average located nearer the platelet surface and we observed extrusion of mitochondria from the platelet surface as microparticles. These data implicate platelet mitochondria in a number of prothrombotic phenomena. Further studies are warranted to assess whether these findings correlate with clinical thrombotic events.
Collapse
Affiliation(s)
- David M Ross
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia.
- Department of Haematology, Royal Adelaide Hospital, 6E359, Port Rd, Adelaide, SA, 5000, Australia.
- Department of Haematology, Flinders University and Medical Centre, Adelaide, Australia.
- Adelaide Medical School, University of Adelaide, Adelaide, Australia.
| | - Hai Po Helena Liang
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia
| | - Zeenet Iqra
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Shane Whittaker
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia
| | - Chuen Wen Tan
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia
| | - Brian J Dale
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Vivien M Chen
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW, Australia
- Department of Haematology, Concord Repatriation General Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, Concord Clinical School, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
19
|
Liu Z, Xu X, Liu K, Zhang J, Ding D, Fu R. Immunogenic Cell Death in Hematological Malignancy Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207475. [PMID: 36815385 PMCID: PMC10161053 DOI: 10.1002/advs.202207475] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/09/2023] [Indexed: 05/06/2023]
Abstract
Although the curative effect of hematological malignancies has been improved in recent years, relapse or drug resistance of hematological malignancies will eventually recur. Furthermore, the microenvironment disorder is an important mechanism in the pathogenesis of hematological malignancies. Immunogenic cell death (ICD) is a unique mechanism of regulated cell death (RCD) that triggers an intact antigen-specific adaptive immune response by firing a set of danger signals or damage-associated molecular patterns (DAMPs), which is an immunotherapeutic modality with the potential for the treatment of hematological malignancies. This review summarizes the existing knowledge about the induction of ICD in hematological malignancies and the current research on combining ICD inducers with other treatment strategies for hematological malignancies.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Xintong Xu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Kaining Liu
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Jingtian Zhang
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Rong Fu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| |
Collapse
|
20
|
Lysenko V, Schürch PM, Tuzlak S, van Wijk NWV, Kovtonyuk LV, Becher B, Manz MG, Kreutmair S, Theocharides APA. Blocking the CD47-SIRPα interaction reverses the disease phenotype in a polycythemia vera mouse model. Leukemia 2023:10.1038/s41375-023-01903-2. [PMID: 37095207 DOI: 10.1038/s41375-023-01903-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/26/2023]
Abstract
Polycythemia vera (PV) is a hematopoietic stem cell neoplasm driven by somatic mutations in JAK2, leading to increased red blood cell (RBC) production uncoupled from mechanisms that regulate physiological erythropoiesis. At steady-state, bone marrow macrophages promote erythroid maturation, whereas splenic macrophages phagocytose aged or damaged RBCs. The binding of the anti-phagocytic ("don't eat me") CD47 ligand expressed on RBCs to the SIRPα receptor on macrophages inhibits phagocytic activity protecting RBCs from phagocytosis. In this study, we explore the role of the CD47-SIRPα interaction on the PV RBC life cycle. Our results show that blocking CD47-SIRPα in a PV mouse model due to either anti-CD47 treatment or loss of the inhibitory SIRPα-signal corrects the polycythemia phenotype. Anti-CD47 treatment marginally impacted PV RBC production while not influencing erythroid maturation. However, upon anti-CD47 treatment, high-parametric single-cell cytometry identified an increase of MerTK+ splenic monocyte-derived effector cells, which differentiate from Ly6Chi monocytes during inflammatory conditions, acquire an inflammatory phagocytic state. Furthermore, in vitro, functional assays showed that splenic JAK2 mutant macrophages were more "pro-phagocytic," suggesting that PV RBCs exploit the CD47-SIRPα interaction to escape innate immune attacks by clonal JAK2 mutant macrophages.
Collapse
Affiliation(s)
- Veronika Lysenko
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Patrick M Schürch
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Selma Tuzlak
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Nicole Wildner-Verhey van Wijk
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Larisa V Kovtonyuk
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Stefanie Kreutmair
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Alexandre P A Theocharides
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Comprehensive Cancer Center Zurich, Zurich, Switzerland.
| |
Collapse
|
21
|
Krecak I, Skelin M, Verstovsek S. Evaluating ropeginterferon alfa-2b for the treatment of adults with polycythemia vera. Expert Rev Hematol 2023; 16:305-316. [PMID: 37002907 DOI: 10.1080/17474086.2023.2199151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
INTRODUCTION Interferons (IFNs) have been used for decades to treat polycythemia vera (PV). Single-arm clinical trials assessing IFN in PV patients demonstrated high hematological and molecular response rates, indicating potential disease-modifying activity of IFN. However, discontinuation rates of IFNs have been rather high due to frequent treatment-related side-effects. AREAS COVERED Ropeginterferon alfa-2b (ROPEG) is a monopegylated IFN consisting of a single isoform, which differentiates it from previous IFNs with respect to tolerability and dosing frequency. ROPEG has improved pharmacokinetic and pharmacodynamic properties, which allow extended dosing every 2 weeks and monthly administration during maintenance phase. This review covers ROPEG's pharmacokinetic and pharmacodynamic properties, presents results of randomized clinical trials (RCT) that evaluated ROPEG in the treatment of PV patients, and discusses contemporary findings regarding the potential disease-modifying activity of ROPEG. EXPERT OPINION RCT have demonstrated high rates of hematological and molecular responses in PV patients treated with ROPEG, irrespective of thrombotic risk. Drug discontinuation rates were generally low. However, even though RCT captured the most important surrogate endpoints of thrombotic risk and disease progression in PV, they were not statistically powered to fully determine whether therapeutic intervention with ROPEG indeed has a direct positive effect on these important clinical outcomes.
Collapse
Affiliation(s)
- Ivan Krecak
- Department of Internal Medicine, General Hospital of Sibenik-Knin County, Sibenik, Croatia
- Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Marko Skelin
- Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Pharmacy Department, General Hospital of Šibenik-Knin County, Šibenik, Croatia
| | - Srdan Verstovsek
- Department of Leukemia, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
22
|
Gotfredsen K, Liisborg C, Skov V, Kjær L, Hasselbalch HC, Sørensen TL. Serum levels of IL-4, IL-13 and IL-33 in patients with age-related macular degeneration and myeloproliferative neoplasms. Sci Rep 2023; 13:4077. [PMID: 36906669 PMCID: PMC10008625 DOI: 10.1038/s41598-023-31078-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/06/2023] [Indexed: 03/13/2023] Open
Abstract
Immune responses play a key role in the pathogenesis and progression of myeloproliferative neoplasms (MPN) and age-related macular degeneration (AMD). Recent studies suggested using MPNs as a "Human Inflammation Model" of drusen development and previous results showed interleukin-4 (IL-4) dysregulation in MPN and AMD. IL-4, IL-13 and IL-33 are all cytokines involved in the type 2 inflammatory response. This study investigated the cytokine levels of IL-4, IL-13 and IL-33 in serum of MPN and AMD patients. This cross-sectional study included 35 patients with MPN with drusen (MPNd) and 27 with MPN and normal retinas (MPNn), 28 patients with intermediate AMD (iAMD) and 29 with neovascular AMD (nAMD). With immunoassays, we quantified and compared levels of IL-4, IL-13 and IL-33 in serum between the groups. The study was conducted at Zealand University Hospital, Roskilde, Denmark, between July 2018 and November 2020. The serum levels of IL-4 were significantly higher in the MPNd group than in the MPNn group (p = 0.003). In regard to IL-33, the difference between MPNd and MPNn was not significant (p = 0.069), however, when subdivided into subgroups, a significant difference was found between polycythemia vera patients with drusen and those without drusen (p = 0.005). We found no IL-13 difference between the MPNd and MPNn groups. Our data didn't show any significant IL-4 or IL-13 serum level difference between the MPNd and iAMD groups but in regard to IL-33, data recorded a significant serum level difference between the two groups. There was no statistically significant difference between the MPNn, iAMD and nAMD groups in levels of IL-4, IL-13 and IL-33. These findings suggested that the serum levels of IL-4 and IL-33 might play a role in drusen development in MPN patients. The results might represent the type 2 inflammatory arm of the disease. The findings support the association between chronic inflammation and drusen.
Collapse
Affiliation(s)
- Kathrine Gotfredsen
- The Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.
| | - Charlotte Liisborg
- The Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark
| | - Vibe Skov
- The Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- The Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | | | | |
Collapse
|
23
|
IL-13/IL-4 signaling contributes to fibrotic progression of the myeloproliferative neoplasms. Blood 2022; 140:2805-2817. [PMID: 36283106 DOI: 10.1182/blood.2022017326] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/05/2022] [Accepted: 10/21/2022] [Indexed: 01/17/2023] Open
Abstract
Myelofibrosis (MF) is a disease associated with high unmet medical needs because allogeneic stem cell transplantation is not an option for most patients, and JAK inhibitors are generally effective for only 2 to 3 years and do not delay disease progression. MF is characterized by dysplastic megakaryocytic hyperplasia and progression to fulminant disease, which is associated with progressively increasing marrow fibrosis. Despite evidence that the inflammatory milieu in MF contributes to disease progression, the specific factors that promote megakaryocyte growth are poorly understood. Here, we analyzed changes in the cytokine profiles of MF mouse models before and after the development of fibrosis, coupled with the analysis of bone marrow populations using single-cell RNA sequencing. We found high interleukin 13 (IL-13) levels in the bone marrow of MF mice. IL-13 promoted the growth of mutant megakaryocytes and induced surface expression of transforming growth factor β and collagen biosynthesis. Similarly, analysis of samples from patients with MF revealed elevated levels of IL-13 in the plasma and increased IL-13 receptor expression in marrow megakaryocytes. In vivo, IL-13 overexpression promoted disease progression, whereas reducing IL-13/IL-4 signaling reduced several features of the disease, including fibrosis. Finally, we observed an increase in the number of marrow T cells and mast cells, which are known sources of IL-13. Together, our data demonstrate that IL-13 is involved in disease progression in MF and that inhibition of the IL-13/IL-4 signaling pathway might serve as a novel therapeutic target to treat MF.
Collapse
|
24
|
Cytokine pathway variants modulate platelet production: IFNA16 is a thrombocytosis susceptibility locus in humans. Blood Adv 2022; 6:4884-4900. [PMID: 35381074 PMCID: PMC9631663 DOI: 10.1182/bloodadvances.2021005648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
Inflammatory stimuli have divergent effects on peripheral platelet counts, although the mechanisms of thrombocytopenic and thrombocytotic responses remain poorly understood. A candidate gene approach targeting 326 polymorphic genes enriched in thrombopoietic and cytokine signaling pathways was applied to identify single nucleotide variants (SNVs) implicated in enhanced platelet responses in cohorts with reactive thrombocytosis (RT) or essential (myeloproliferative neoplasm [MPN]) thrombocytosis (ET). Cytokine profiles incorporating a 15-member subset, pathway topology, and functional interactive networks were distinct between ET and RT, consistent with distinct regulatory pathways of exaggerated thrombopoiesis. Genetic studies using aggregate (ET + RT) or ET-restricted cohorts identified associations with 2 IFNA16 (interferon-α16) SNVs, and the ET associations were validated in a second independent cohort (P = .0002). Odds ratio of the combined ET cohort (n = 105) was 4.92, restricted to the JAK2V617F-negative subset (odds ratio, 5.01). ET substratification analysis by variant IFNA16 exhibited a statistically significant increase in IFN-α16 levels (P = .002) among 16 quantifiable cytokines. Recombinantly expressed variant IFN-α16 encompassing 3 linked non-synonymous SNVs (E65H95P133) retained comparable antiviral and pSTAT signaling profiles as native IFN-α16 (V65D95A133) or IFN-α2, although both native and variant IFN-α16 showed stage-restricted differences (compared with IFN-α2) of IFN-regulated genes in CD34+-stimulated megakaryocytes. These data implicate IFNA16 (IFN-α16 gene product) as a putative susceptibility locus (driver) within the broader disrupted cytokine network evident in MPNs, and they provide a framework for dissecting functional interactive networks regulating stress or MPN thrombopoiesis.
Collapse
|
25
|
Leiva O, Hobbs G, Ravid K, Libby P. Cardiovascular Disease in Myeloproliferative Neoplasms: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2022; 4:166-182. [PMID: 35818539 PMCID: PMC9270630 DOI: 10.1016/j.jaccao.2022.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 11/24/2022] Open
Abstract
Myeloproliferative neoplasms are associated with increased risk for thrombotic complications. These conditions most commonly involve somatic mutations in genes that lead to constitutive activation of the Janus-associated kinase signaling pathway (eg, Janus kinase 2, calreticulin, myeloproliferative leukemia protein). Acquired gain-of-function mutations in these genes, particularly Janus kinase 2, can cause a spectrum of disorders, ranging from clonal hematopoiesis of indeterminate potential, a recently recognized age-related promoter of cardiovascular disease, to frank hematologic malignancy. Beyond thrombosis, patients with myeloproliferative neoplasms can develop other cardiovascular conditions, including heart failure and pulmonary hypertension. The authors review the pathophysiologic mechanisms of cardiovascular complications of myeloproliferative neoplasms, which involve inflammation, prothrombotic and profibrotic factors (including transforming growth factor-beta and lysyl oxidase), and abnormal function of circulating clones of mutated leukocytes and platelets from affected individuals. Anti-inflammatory therapies may provide cardiovascular benefit in patients with myeloproliferative neoplasms, a hypothesis that requires rigorous evaluation in clinical trials.
Collapse
Key Words
- ASXL1, additional sex Combs-like 1
- CHIP, clonal hematopoiesis of indeterminate potential
- DNMT3a, DNA methyltransferase 3 alpha
- IL, interleukin
- JAK, Janus-associated kinase
- JAK2, Janus kinase 2
- LOX, lysyl oxidase
- MPL, myeloproliferative leukemia protein
- MPN, myeloproliferative neoplasm
- STAT, signal transducer and activator of transcription
- TET2, tet methylcytosine dioxygenase 2
- TGF, transforming growth factor
- atherosclerosis
- cardiovascular complications
- clonal hematopoiesis
- myeloproliferative neoplasms
- thrombosis
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gabriela Hobbs
- Division of Hematology Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Wang JC, Sun L. PD-1/PD-L1, MDSC Pathways, and Checkpoint Inhibitor Therapy in Ph(-) Myeloproliferative Neoplasm: A Review. Int J Mol Sci 2022; 23:5837. [PMID: 35628647 PMCID: PMC9143160 DOI: 10.3390/ijms23105837] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/23/2022] Open
Abstract
There has been significant progress in immune checkpoint inhibitor (CPI) therapy in many solid tumor types. However, only a single failed study has been published in treating Ph(-) myeloproliferative neoplasm (MPN). To make progress in CPI studies on this disease, herein, we review and summarize the mechanisms of activation of the PD-L1 promoter, which are as follows: (a) the extrinsic mechanism, which is activated by interferon gamma (IFN γ) by tumor infiltration lymphocytes (TIL) and NK cells; (b) the intrinsic mechanism of EGFR or PTEN loss resulting in the activation of the MAPK and AKT pathways and then stat 1 and 3 activation; and (c) 9p24 amplicon amplification, resulting in PD-L1 and Jak2 activation. We also review the literature and postulate that many of the failures of CPI therapy in MPN are likely due to excessive MDSC activities. We list all of the anti-MDSC agents, especially those with ruxolitinib, IMID compounds, and BTK inhibitors, which may be combined with CPI therapy in the future as part of clinical trials applying CPI therapy to Ph(-) MPN.
Collapse
Affiliation(s)
- Jen-Chin Wang
- Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY 11212, USA;
| | | |
Collapse
|
27
|
Carnaz Simões AM, Holmström MO, Aehnlich P, Rahbech A, Peeters MJW, Radziwon-Balicka A, Zamora C, Wirenfeldt Klausen T, Skov V, Kjær L, Ellervik C, Fassi DE, Vidal S, Hasselbalch HC, Andersen MH, thor Straten P. Patients With Myeloproliferative Neoplasms Harbor High Frequencies of CD8 T Cell-Platelet Aggregates Associated With T Cell Suppression. Front Immunol 2022; 13:866610. [PMID: 35603202 PMCID: PMC9120544 DOI: 10.3389/fimmu.2022.866610] [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: 01/31/2022] [Accepted: 04/07/2022] [Indexed: 11/16/2022] Open
Abstract
Myeloproliferative neoplasms (MPN) are chronic cancers of the hematopoietic stem cells in the bone marrow, and patients often harbor elevated numbers of circulating platelets (PLT). We investigated the frequencies of circulating PLT-lymphocyte aggregates in MPN patients and the effect of PLT-binding on CD8 T cell function. The phenotype of these aggregates was evaluated in 50 MPN patients and 24 controls, using flow cytometry. In vitro studies compared the proliferation, cytokine release, and cytoxicity of PLT-bound and PLT-free CD8 T cells. Frequencies of PLT-CD8 T cell aggregates, were significantly elevated in MPN patients. Advanced disease stage and CALR mutation associated with the highest aggregate frequencies with a predominance of PLT-binding to antigen-experienced CD8 T cells. PLT-bound CD8 T cells showed reduction in proliferation and cytotoxic capacity. Our data suggest that CD8 T cell responses are jeopardized in MPN patients. JAK2 and CALR exon 9 mutations - the two predominant driver mutations in MPN - are targets for natural T cell responses in MPN patients. Moreover, MPN patients have more infections compared to background. Thus, PLT binding to antigen experienced CD8 T cells could play a role in the inadequacy of the immune system to control MPN disease progression and prevent recurrent infections.
Collapse
Affiliation(s)
- Ana Micaela Carnaz Simões
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Herlev University Hospital, Herlev, Denmark
| | - Morten Orebo Holmström
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Herlev University Hospital, Herlev, Denmark
| | - Pia Aehnlich
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Herlev University Hospital, Herlev, Denmark
| | - Anne Rahbech
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Herlev University Hospital, Herlev, Denmark
| | - Marlies J. W. Peeters
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Herlev University Hospital, Herlev, Denmark
| | - Aneta Radziwon-Balicka
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Herlev University Hospital, Herlev, Denmark
| | - Carlos Zamora
- IIB-Sant Pau- Institut Rec. Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Tobias Wirenfeldt Klausen
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), 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
| | - Christina Ellervik
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Laboratory Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Data and Innovation Support, Region Zealand, Sorø, Denmark
| | - Daniel El Fassi
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Hematology, Rigshospitalet University Hospital, Copenhagen, Denmark
| | - Silvia Vidal
- IIB-Sant Pau- Institut Rec. Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Mads Hald Andersen
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Herlev University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Per thor Straten
- Department of Oncology, National Center for Cancer Immune Therapy (CCIT-DK), Herlev University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Alvarez-Larrán A, Garrote M, Ferrer-Marín F, Pérez-Encinas M, Mata-Vazquez MI, Bellosillo B, Arellano-Rodrigo E, Gómez M, García R, García-Gutiérrez V, Gasior M, Cuevas B, Angona A, Gómez-Casares MT, Martínez CM, Magro E, Ayala R, Del Orbe-Barreto R, Pérez-López R, Fox ML, Raya JM, Guerrero L, García-Hernández C, Caballero G, Murillo I, Xicoy B, Ramírez MJ, Carreño-Tarragona G, Hernández-Boluda JC, Pereira A. Real-world analysis of main clinical outcomes in patients with polycythemia vera treated with ruxolitinib or best available therapy after developing resistance/intolerance to hydroxyurea. Cancer 2022; 128:2441-2448. [PMID: 35417564 PMCID: PMC9324831 DOI: 10.1002/cncr.34195] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/21/2022] [Accepted: 02/14/2022] [Indexed: 11/16/2022]
Abstract
Background Ruxolitinib is approved for patients with polycythemia vera (PV) who are resistant/intolerant to hydroxyurea, but its impact on preventing thrombosis or disease‐progression is unknown. Methods A retrospective, real‐world analysis was performed on the outcomes of 377 patients with resistance/intolerance to hydroxyurea from the Spanish Registry of Polycythemia Vera according to subsequent treatment with ruxolitinib (n = 105) or the best available therapy (BAT; n = 272). Survival probabilities and rates of thrombosis, hemorrhage, acute myeloid leukemia, myelofibrosis, and second primary cancers were calculated according to treatment. To minimize biases in treatment allocation, all results were adjusted by a propensity score for receiving ruxolitinib or BAT. Results Patients receiving ruxolitinib had a significantly lower rate of arterial thrombosis than those on BAT (0.4% vs 2.3% per year; P = .03), and this persisted as a trend after adjustment for the propensity to have received the drug (incidence rate ratio, 0.18; 95% confidence interval, 0.02‐1.3; P = .09). There were no significant differences in the rates of venous thrombosis (0.8% and 1.1% for ruxolitinib and BAT, respectively; P = .7) and major bleeding (0.8% and 0.9%, respectively; P = .9). Ruxolitinib exposure was not associated with a higher rate of second primary cancers, including all types of neoplasia, noncutaneous cancers, and nonmelanoma skin cancers. After a median follow‐up of 3.5 years, there were no differences in survival or progression to acute leukemia or myelofibrosis between the 2 groups. Conclusions The results suggest that ruxolitinib treatment for PV patients with resistance/intolerance to hydroxyurea may reduce the incidence of arterial thrombosis. Lay Summary Ruxolitinib is better than other available therapies in achieving hematocrit control and symptom relief in patients with polycythemia vera who are resistant/intolerant to hydroxyurea, but we still do not know whether ruxolitinib provides an additional benefit in preventing thrombosis or disease progression. We retrospectively studied the outcomes of 377 patients with resistance/intolerance to hydroxyurea from the Spanish Registry of Polycythemia Vera according to whether they subsequently received ruxolitinib (n = 105) or the best available therapy (n = 272). Our findings suggest that ruxolitinib could reduce the incidence of arterial thrombosis, but a disease‐modifying effect could not be demonstrated for ruxolitinib in this patient population.
Ruxolitinib may reduce the incidence of arterial thrombosis in patients with polycythemia vera resistant/intolerant to hydroxyurea.
Collapse
Affiliation(s)
- Alberto Alvarez-Larrán
- Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Marta Garrote
- Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Francisca Ferrer-Marín
- Hospital Morales Messeguer, Universidad Católica San Antonio de Murcia, Murcia, Centro de Investigación Biomédica en Red de Enfermedades Raras, Murcia, Spain
| | | | | | | | | | | | | | | | | | | | - Anna Angona
- Hospital Josep Trueta, Institut Català d'Oncologia, Girona, Spain
| | | | | | - Elena Magro
- Hospital Príncipe de Asturias, Alcalá de Henares, Spain
| | - Rosa Ayala
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | | | - José-María Raya
- Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | | | | | | | | | - Blanca Xicoy
- Hospital Germans Trias i Pujol, Institut Català d'Oncologia, Josep Carreras Leukemia Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| | | | | | | | - Arturo Pereira
- Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | |
Collapse
|
29
|
Bhuria V, Baldauf CK, Schraven B, Fischer T. Thromboinflammation in Myeloproliferative Neoplasms (MPN)-A Puzzle Still to Be Solved. Int J Mol Sci 2022; 23:ijms23063206. [PMID: 35328626 PMCID: PMC8954909 DOI: 10.3390/ijms23063206] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs), a group of malignant hematological disorders, occur as a consequence of somatic mutations in the hematopoietic stem cell compartment and show excessive accumulation of mature myeloid cells in the blood. A major cause of morbidity and mortality in these patients is the marked prothrombotic state leading to venous and arterial thrombosis, including myocardial infarction (MI), deep vein thrombosis (DVT), and strokes. Additionally, many MPN patients suffer from inflammation-mediated constitutional symptoms, such as fever, night sweats, fatigue, and cachexia. The chronic inflammatory syndrome in MPNs is associated with the up-regulation of various inflammatory cytokines in patients and is involved in the formation of the so-called MPN thromboinflammation. JAK2-V617F, the most prevalent mutation in MPNs, has been shown to activate a number of integrins on mature myeloid cells, including granulocytes and erythrocytes, which increase adhesion and drive venous thrombosis in murine knock-in/out models. This review aims to shed light on the current understanding of thromboinflammation, involvement of neutrophils in the prothrombotic state, plausible molecular mechanisms triggering the process of thrombosis, and potential novel therapeutic targets for developing effective strategies to reduce the MPN disease burden.
Collapse
Affiliation(s)
- Vikas Bhuria
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Conny K. Baldauf
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-67-15338; Fax: +49-391-67-15852
| | - Thomas Fischer
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
30
|
Bhuria V, Baldauf CK, Schraven B, Fischer T. Thromboinflammation in Myeloproliferative Neoplasms (MPN)-A Puzzle Still to Be Solved. Int J Mol Sci 2022. [PMID: 35328626 DOI: 10.3390/ijms23063206.pmid:35328626;pmcid:pmc8954909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs), a group of malignant hematological disorders, occur as a consequence of somatic mutations in the hematopoietic stem cell compartment and show excessive accumulation of mature myeloid cells in the blood. A major cause of morbidity and mortality in these patients is the marked prothrombotic state leading to venous and arterial thrombosis, including myocardial infarction (MI), deep vein thrombosis (DVT), and strokes. Additionally, many MPN patients suffer from inflammation-mediated constitutional symptoms, such as fever, night sweats, fatigue, and cachexia. The chronic inflammatory syndrome in MPNs is associated with the up-regulation of various inflammatory cytokines in patients and is involved in the formation of the so-called MPN thromboinflammation. JAK2-V617F, the most prevalent mutation in MPNs, has been shown to activate a number of integrins on mature myeloid cells, including granulocytes and erythrocytes, which increase adhesion and drive venous thrombosis in murine knock-in/out models. This review aims to shed light on the current understanding of thromboinflammation, involvement of neutrophils in the prothrombotic state, plausible molecular mechanisms triggering the process of thrombosis, and potential novel therapeutic targets for developing effective strategies to reduce the MPN disease burden.
Collapse
Affiliation(s)
- Vikas Bhuria
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Conny K Baldauf
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Thomas Fischer
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
31
|
Hodeib H, Abd EL Hai D, Tawfik MA, Allam AA, Selim A, Elsawy AA, Youssef A. CCL2 rs1024611Gene Polymorphism in Philadelphia-Negative Myeloproliferative Neoplasms. Genes (Basel) 2022; 13:genes13030492. [PMID: 35328046 PMCID: PMC8948730 DOI: 10.3390/genes13030492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/20/2022] [Accepted: 03/08/2022] [Indexed: 02/05/2023] Open
Abstract
Introduction: The onset of the Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs) is caused by acquired somatic mutations in target myeloid genes “driver mutations”. The CCL2 gene is overexpressed by non-Hodgkin lymphomas and multiple solid tumors. Aim of the study: to evaluate the possible association of CCL2 rs1024611 SNP and its expression level and the risk of developing Philadelphia-negative MPNs. Patients and methods: A total of 128 newly diagnosed Philadelphia-negative MPN patient and 141 healthy subjects were evaluated for the genotype distribution of CCL2 rs1024611 and CCL2 expression levels. Results: The CCL2 rs1024611 G/G genotype was more frequent and significantly frequent among PMF and Post-PV/ET-MF patients and the mean CCL2 expression levels were significantly higher in PMF and Post-PV/ET-MF compared to the healthy subjects. The CCL2 rs1024611 SNP was significantly correlated to the CCL2 gene expression level and fibrosis grade. ROC analysis for the CCL2 gene expression level that discriminates MF patients from PV + ET patients revealed a sensitivity of 80.43% and a specificity of 73.17% with an AUC of 0.919 (p < 0.001). Conclusion: The CCL2 rs1024611 polymorphism could be an independent risk factor for developing MF (PMF and Post-PV/ET-MF). Moreover, CCL2 gene expression could be potential genetic biomarker of fibrotic progression.
Collapse
Affiliation(s)
- Hossam Hodeib
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (H.H.); (D.A.E.H.); (A.Y.)
| | - Dina Abd EL Hai
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (H.H.); (D.A.E.H.); (A.Y.)
| | - Mohamed A Tawfik
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (A.A.A.); (A.S.); (A.A.E.)
- Correspondence: ; Tel.: +20-1550844075
| | - Alzahraa A. Allam
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (A.A.A.); (A.S.); (A.A.E.)
| | - Amal Selim
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (A.A.A.); (A.S.); (A.A.E.)
| | - Abdallah Ahmed Elsawy
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (A.A.A.); (A.S.); (A.A.E.)
| | - Amira Youssef
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (H.H.); (D.A.E.H.); (A.Y.)
| |
Collapse
|
32
|
Strickland M, Quek L, Psaila B. The immune landscape in BCR-ABL negative myeloproliferative neoplasms: inflammation, infections and opportunities for immunotherapy. Br J Haematol 2022; 196:1149-1158. [PMID: 34618358 PMCID: PMC9135025 DOI: 10.1111/bjh.17850] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 09/05/2021] [Accepted: 09/11/2021] [Indexed: 01/06/2023]
Abstract
Breakpoint cluster region-Abelson (BCR-ABL) negative myeloproliferative neoplasms (MPNs) are chronic myeloid neoplasms initiated by the acquisition of gene mutation(s) in a haematopoietic stem cell, leading to clonal expansion and over-production of blood cells and their progenitors. MPNs encompass a spectrum of disorders with overlapping but distinct molecular, laboratory and clinical features. This includes polycythaemia vera, essential thrombocythaemia and myelofibrosis. Dysregulation of the immune system is key to the pathology of MPNs, supporting clonal evolution, mediating symptoms and resulting in varying degrees of immunocompromise. Targeting immune dysfunction is an important treatment strategy. In the present review, we focus on the immune landscape in patients with MPNs - the role of inflammation in disease pathogenesis, susceptibility to infection and emerging strategies for therapeutic immune modulation. Further detailed work is required to delineate immune perturbation more precisely in MPNs to determine how and why vulnerability to infection differs between clinical subtypes and to better understand how inflammation results in a competitive advantage for the MPN clone. These studies may help shed light on new designs for disease-modifying therapies.
Collapse
Affiliation(s)
- Marie Strickland
- MRC Molecular Haematology UnitMRC Weatherall Institute of Molecular Medicine, University of OxfordOxford
- National Institutes for Health Research Biomedical Research CentreUniversity of OxfordOxford
| | - Lynn Quek
- Department of Haematological MedicineKing's College Hospital NHS Foundation TrustLondon
- Department of Haematology, School of Cancer and Pharmaceutical SciencesKing's College LondonLondonUK
| | - Bethan Psaila
- MRC Molecular Haematology UnitMRC Weatherall Institute of Molecular Medicine, University of OxfordOxford
- National Institutes for Health Research Biomedical Research CentreUniversity of OxfordOxford
| |
Collapse
|
33
|
Sobas M, Podolak-Dawidziak M, Lewandowski K, Bator M, Wróbel T. Primary Immune Thrombocytopenia and Essential Thrombocythemia: So Different and yet Somehow Similar-Cases Series and a Review of the Literature. Int J Mol Sci 2021; 22:10918. [PMID: 34681577 PMCID: PMC8539407 DOI: 10.3390/ijms222010918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/02/2021] [Accepted: 10/06/2021] [Indexed: 12/02/2022] Open
Abstract
This article collects several published cases in which immune thrombocytopenic purpura (ITP) is followed by essential thrombocythemia (ET) and vice versa. This surprising clinical condition is possible, but very rare and difficult to diagnose and manage. We have made an attempt to analyse the possible causes of the sequential appearance of ITP and ET taking into consideration the following: alteration of the thrombopoietin (TPO) receptor, the role of autoimmunity and inflammation, and cytokine modulation. A better understanding of these interactions may provide opportunities to determine predisposing factors and aid in finding new treatment modalities both for ITP and ET patients.
Collapse
Affiliation(s)
- Marta Sobas
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Pasteura 4, 50-367 Wroclaw, Poland; (M.P.-D.); (M.B.); (T.W.)
| | - Maria Podolak-Dawidziak
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Pasteura 4, 50-367 Wroclaw, Poland; (M.P.-D.); (M.B.); (T.W.)
| | - Krzysztof Lewandowski
- Hematology and Bone Marrow Transplantation Department, University of Medical Sciences, 60-569 Poznan, Poland;
| | - Michał Bator
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Pasteura 4, 50-367 Wroclaw, Poland; (M.P.-D.); (M.B.); (T.W.)
| | - Tomasz Wróbel
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Pasteura 4, 50-367 Wroclaw, Poland; (M.P.-D.); (M.B.); (T.W.)
| |
Collapse
|
34
|
Anti-inflammatory treatment in MPN: Targeting TNFα-receptor 1 (TNFR1) and TNFR2 in JAK2-V617F induced disease. Blood Adv 2021; 5:5349-5359. [PMID: 34592754 PMCID: PMC9153051 DOI: 10.1182/bloodadvances.2021004438] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022] Open
Abstract
Inhibition of TNFR2 decreases WBC counts but does not ameliorate hematocrit and splenomegaly in a JAK2-V617F knock-in mouse model. In a JAK2-V617F knock-in mouse model expressing chimeric TNFR1, anti-human TNFR1 antibody therapy reduces hematocrit and splenomegaly.
Chronic nonresolving inflammatory syndrome is a major disease feature in myeloproliferative neoplasms (MPNs). Systemic inflammation promotes the growth of the JAK2-V617F+ hematopoietic stem cell clone and is associated with constitutive symptoms (eg, fever, cachexia, and fatigue). Therefore, it is being discussed whether anti-inflammatory therapy, in addition to the well-established JAK inhibitor therapy, may be beneficial in the control of constitutive symptoms. Moreover, effective control of the inflammatory microenvironment may contribute to prevent transformation into secondary myelofibrosis and acute leukemia. Given the pivotal role of tumor necrosis factor α (TNF-α) in MPN and the distinct roles of TNF-α receptor 1 (TNFR1) and TNFR2 in inflammation, we investigated the therapeutic effects of αTNFR1 and αTNFR2 antibody treatment in MPN-like disease using the JAK2+/VF knock-in mouse model. Peripheral blood counts, bone marrow/spleen histopathology, and inflammatory cytokine levels in serum were investigated. αTNFR2 antibody treatment decreased white blood cells and modulated the serum levels of several cytokines [CXCL2, CXCL5, interleukin-12(p40)], as well as of macrophage colony-stimulating factor, but they lacked efficacy to ameliorate hematocrit and splenomegaly. αTNFR1 antibody treatment resulted in the mild suppression of elevated hematocrit of −10.7% and attenuated splenomegaly (22% reduction in spleen weight). In conclusion, our studies show that TNFR1 and TNFR2 play different roles in the biology of JAK2-V617F–induced disease that may be of relevance in future therapeutic settings.
Collapse
|
35
|
Coltro G, Loscocco GG, Vannucchi AM. Classical Philadelphia-negative myeloproliferative neoplasms (MPNs): A continuum of different disease entities. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 365:1-69. [PMID: 34756241 DOI: 10.1016/bs.ircmb.2021.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Classical Philadelphia-negative myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell-derived disorders characterized by uncontrolled proliferation of differentiated myeloid cells and close pathobiologic and clinical features. According to the 2016 World Health Organization (WHO) classification, MPNs include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). The 2016 revision aimed in particular at strengthening the distinction between masked PV and JAK2-mutated ET, and between prefibrotic/early (pre-PMF) and overt PMF. Clinical manifestations in MPNs include constitutional symptoms, microvascular disorders, thrombosis and bleeding, splenomegaly secondary to extramedullary hematopoiesis, cytopenia-related symptoms, and progression to overt MF and acute leukemia. A dysregulation of the JAK/STAT pathway is the unifying mechanistic hallmark of MPNs, and is guided by somatic mutations in driver genes including JAK2, CALR and MPL. Additional mutations in myeloid neoplasm-associated genes have been also identified, with established prognostic relevance, particularly in PMF. Prognostication of MPN patients relies on disease-specific clinical models. The increasing knowledge of MPN biology led to the development of integrated clinical and molecular prognostic scores that allow a more refined stratification. Recently, the therapeutic landscape of MPNs has been revolutionized by the introduction of potent, selective JAK inhibitors (ruxolitinib, fedratinib), that proved effective in controlling disease-related symptoms and splenomegaly, yet leaving unmet critical needs, owing the lack of disease-modifying activity. In this review, we will deal with molecular, clinical, and therapeutic aspects of the three classical MPNs aiming at highlighting either shared characteristics, that overall define a continuum within a single disease family, and uniqueness, at the same time.
Collapse
Affiliation(s)
- Giacomo Coltro
- CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giuseppe G Loscocco
- CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro M Vannucchi
- CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
36
|
Anti-IL-6 cytokine treatment has no impact on elevated hematocrit and splenomegaly in a polycythemia vera mouse model. Blood Adv 2021; 6:399-404. [PMID: 34559181 PMCID: PMC8791576 DOI: 10.1182/bloodadvances.2021004379] [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: 01/28/2021] [Accepted: 07/26/2021] [Indexed: 11/20/2022] Open
Abstract
Somatic mutations in JAK2, MPL and Calreticulin and inflammation play a key role in pathophysiology of chronic myeloproliferative neoplasia (CMN). One of the most prominent cytokines elevated in serum of Polycythemia vera patients is interleukin-6 (IL-6). Currently, it is being discussed whether suppression of inflammation by anti-cytokine approaches as anti-IL-6 treatment may be therapeutically useful in CMN. We here sought to investigate the efficacy of anti-IL-6 treatment on inflammatory cytokines, hematocrit and splenomegaly in CMN like disease. JAK2-V617F knock-in mice (JAK2+/V617F) were treated for three weeks with anti-IL-6 antibody (Ab) or IgG-control. Upon anti-IL-6 Ab treatment, serum levels of CXCL2 and CXCL10 were significantly reduced. In addition, CXCL1, CCL11, M-CSF, G-CSF, IL-17, IL-12p40 and CCL2 were reduced by a factor of 0.3 - 0.8. Partly, this was also achieved by applying high-dose IgG. Hematocrit, erythrocyte and leukocyte counts were elevated in JAK2+/V617F mice but were not reduced by anti-IL6 Ab treatment. In addition, there was no apparent amelioration of splenomegaly and spleen histopathology. In conclusion, anti-IL-6 Ab treatment did not result in improvement of hematological disease parameters but was shown to modulate the serum cytokine signature.
Collapse
|
37
|
The Power of Extracellular Vesicles in Myeloproliferative Neoplasms: "Crafting" a Microenvironment That Matters. Cells 2021; 10:cells10092316. [PMID: 34571965 PMCID: PMC8464728 DOI: 10.3390/cells10092316] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022] Open
Abstract
Myeloproliferative Neoplasms (MPN) are acquired clonal disorders of the hematopoietic stem cells and include Essential Thrombocythemia, Polycythemia Vera and Myelofibrosis. MPN are characterized by mutations in three driver genes (JAK2, CALR and MPL) and by a state of chronic inflammation. Notably, MPN patients experience increased risk of thrombosis, disease progression, second neoplasia and evolution to acute leukemia. Extracellular vesicles (EVs) are a heterogeneous population of microparticles with a role in cell-cell communication. The EV-mediated cross-talk occurs via the trafficking of bioactive molecules such as nucleic acids, proteins, metabolites and lipids. Growing interest is focused on EVs and their potential impact on the regulation of blood cancers. Overall, EVs have been suggested to orchestrate the complex interplay between tumor cells and the microenvironment with a pivotal role in "education" and "crafting" of the microenvironment by regulating angiogenesis, coagulation, immune escape and drug resistance of tumors. This review is focused on the role of EVs in MPN. Specifically, we will provide an overview of recent findings on the involvement of EVs in MPN pathogenesis and discuss opportunities for their potential application as diagnostic and prognostic biomarkers.
Collapse
|
38
|
How J, Hobbs G. Management Issues and Controversies in Low-Risk Patients with Essential Thrombocythemia and Polycythemia Vera. Curr Hematol Malig Rep 2021; 16:473-482. [PMID: 34478054 DOI: 10.1007/s11899-021-00649-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Essential thrombocythemia (ET) and polycythemia vera (PV) are the most common myeloproliferative neoplasms (MPNs). Treatment of ET and PV is based on the risk for subsequent thrombosis. High-risk patients, defined as older than 60, JAK2 V617F-positive patients, or patients with a history of prior thrombosis, merit cytoreduction to control blood counts, whereas a watchful waiting paradigm is utilized in low-risk patients. However, low-risk patients have a host of other specific management issues that arise during their disease course. This review will discuss the most common management issues specific to the care of low-risk patients, including anti-platelet therapy dosing, pregnancy, and indications for early cytoreduction. RECENT FINDINGS Although low-dose aspirin is well established in PV, its indications and dosing regimens are less clear in ET. Recent evidence has supported twice daily low-dose aspirin in ET and observation alone in very low-risk ET patients. Pregnancy is not contraindicated in MPNs, and we recommend aspirin throughout pregnancy with consideration for prophylactic postpartum anticoagulation. High phlebotomy needs, symptom burden, and extreme thrombocytosis are common reasons for initiation of cytoreduction in low-risk patients, although we typically do not start cytoreduction for an isolated high platelet count alone. Recent data has also demonstrated a potential disease-modifying effect of interferons in MPNs, with some experts now advocating the early use of interferon in low-risk patients, although more mature data is needed before practice guidelines change. We evaluate the literature to inform clinical decision-making regarding these controversies, including most recent data that has challenged the "watchful waiting" paradigm. Our discussion provides guidance on common clinical scenarios seen in low-risk ET and PV patients, who face a myriad of complex management decisions in their care.
Collapse
Affiliation(s)
- Joan How
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Zero Emerson, Office 138, Boston, MA, 02114, USA.,Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Gabriela Hobbs
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Zero Emerson, Office 138, Boston, MA, 02114, USA.
| |
Collapse
|
39
|
Myeloproliferative Disorders and its Effect on Bone Homeostasis: The Role of Megakaryocytes. Blood 2021; 139:3127-3137. [PMID: 34428274 DOI: 10.1182/blood.2021011480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/11/2021] [Indexed: 11/20/2022] Open
Abstract
Myeloproliferative Neoplasms (MPNs) are a heterogeneous group of chronic hematological diseases that arise from the clonal expansion of abnormal hematopoietic stem cells, of which Polycythemia Vera (PV), Essential Thrombocythemia (ET), and Primary Myelofibrosis (PMF) have been extensively reviewed in context of clonal expansion, fibrosis and other phenotypes. Here, we review current knowledge on the influence of different forms of MPN on bone health. Studies implicated various degrees of effect of different forms of MPN on bone density, and on osteoblast proliferation and differentiation, using murine models and human data. The majority of studies show that bone volume is generally increased in PMF patients, whereas it is slightly decreased or not altered in ET and PV patients, although possible differences between male and female phenotypes were not fully explored in most MPN forms. Osteosclerosis seen in PMF patients is a serious complication that can lead to bone marrow failure, and the loss of bone reported in some ET and PV patients can lead to osteoporotic fractures. Some MPN forms are associated with increased number of megakaryocytes (MKs), and several of the MK-associated factors in MPN are known to affect bone development. Here, we review known mechanisms involved in these processes, with focus on the role of MKs and secreted factors. Understanding MPN-associated changes in bone health could improve early intervention and treatment of this side effect of the pathology.
Collapse
|
40
|
Hui W, Zhang W, Liu C, Wan S, Sun W, Su L. Alterations of Signaling Pathways in Essential Thrombocythemia with Calreticulin Mutation. Cancer Manag Res 2021; 13:6231-6238. [PMID: 34393515 PMCID: PMC8357313 DOI: 10.2147/cmar.s316919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022] Open
Abstract
Purpose Though mutations of the calreticulin (CALR) gene have been identified in essential thrombocythemia patients, the detailed mechanisms for CALR mutations have not been completely clarified. Our study is aimed at characterizing alteration of protein expression in ET patients with mutated CALRdel52 and further recognizing possible involvement of signaling pathways associated with CALR mutations. Patients and Methods Protein pathway array was performed to analyze the expression levels of proteins involved in various signaling pathways in peripheral blood neutrophils from 18 ET patients with mutated CALRdel52, 20 ET patients with JAK2V617F mutation and 20 controls. Results We found 20 proteins differentially expressed in ET patients with mutated CALRdel52 compared with healthy controls. These proteins were associated with molecular mechanisms of cancer in ingenuity pathways analysis (IPA) network. We identified top ten canonical pathways which including apoptotic pathways and cellular cytokine pathways might participate in pathogenesis of ET with mutated CALRdel52. Additionally, there were 8 proteins found to be dysregulated differently between ET patients with mutated CALRdel52 and those with JAK2V617F mutation. These proteins might be related to the unique signaling pathways activated by CALRdel52 mutation which were different to JAK/STATs pathway by JAK2V617F mutation. Conclusion Our study demonstrated that numerous alterations of signaling proteins and pathways in ET patients with mutated CALRdel52. These findings could help to gain insights into the pathological mechanisms of ET.
Collapse
Affiliation(s)
- Wuhan Hui
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wei Zhang
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Congyan Liu
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Suigui Wan
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wanling Sun
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Li Su
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
41
|
Chen P, Wu B, Ji L, Zhan Y, Li F, Cheng L, Cao J, Chen H, Ke Y, Min Z, Sun L, Hua F, Chen H, Cheng Y. Cytokine Consistency Between Bone Marrow and Peripheral Blood in Patients With Philadelphia-Negative Myeloproliferative Neoplasms. Front Med (Lausanne) 2021; 8:598182. [PMID: 34249954 PMCID: PMC8264196 DOI: 10.3389/fmed.2021.598182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 04/09/2021] [Indexed: 12/23/2022] Open
Abstract
Background: Inflammation might play a critical role in the pathogenesis and progression of Philadelphia-negative myeloproliferative neoplasms (Ph−MPNs) with elevated inflammatory cytokines in peripheral blood (PB). However, the inflammatory status inside the bone marrow (BM), which is the place of malignancy origin and important microenvironment of neoplasm evolution, has not yet been elucidated. Methods: Inflammatory cytokine profiles in PB and BM of 24 Ph-MPNs patients were measured by a multiplex quantitative inflammation array. Cytokines that correlated between PB and BM were selected and then validated by ELISA in a separate cohort of 52 MPN patients. Furthermore, a panel of cytokines was identified and examined for potential application as non-invasive markers for the diagnosis and prediction of fibrosis progress of MPN subtypes. Results: The levels of G-CSF, I-309, IL-1β, IL-1ra, IL-12p40, IL-15, IL-16, M-CSF, MIG, PDGF-BB, and TIMP-1 in BM supernatants were significantly higher than those in PB (all p < 0.05). Linear correlations between BM and PB levels were found in 13 cytokines, including BLC, Eotaxin-2, I-309, sICAM-1, IL-15, M-CSF, MIP-1α, MIP-1δ, RANTES, TIMP-1, TIMP-2, sTNFRI, and sTNFRII (all R > 0.4 and p < 0.05). Levels of BLC, Eotaxin-2, M-CSF, and TIMP-1 in PB were significantly different from those in health controls (all p < 0.05). In PB, levels of TIMP-1 and Eotaxin-2 in essential thrombocythemia (ET) group were significantly lower than those in groups of prefibrotic primary myelofibrosis (pre-PMF) [TIMP-1: 685.2 (322.2–1,229) ng/ml vs. 1,369 (1,175–1,497) ng/ml, p = 0.0221; Eotaxin-2: 531.4 (317.9–756.6) pg/ml vs. 942.4 (699.3–1,474) pg/ml, p = 0.0393] and primary myelofibrosis (PMF) [TIMP-1: 685.2 (322.2–1229) ng/ml vs. 1,365 (1,115–1,681) ng/ml, p = 0.0043; Eotaxin-2: 531.4 (317.9–756.6) pg/ml vs. 1,010 (818–1,556) pg/ml, p = 0.0030]. The level of TIMP-1 in myelofibrosis (MF) >1 group was significantly higher than that in MF ≤ 1 group. Conclusion: Abnormal inflammatory status is present in MPN, especially in its BM microenvironment. Consistency between PB and BM levels was found in multiple inflammatory cytokines. Circulating cytokine levels of BLC, M-CSF, Eotaxin-2, and TIMP-1 reflected inflammation inside BM niche, suggesting potential diagnostic value for MPN subtypes and prognostic value for fibrosis progression.
Collapse
Affiliation(s)
- Pu Chen
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Boting Wu
- Department of Transfusion Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lili Ji
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanxia Zhan
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Feng Li
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Hematology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai, China
| | - Luya Cheng
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingjing Cao
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China.,Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hehui Chen
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Ke
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihui Min
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lihua Sun
- Department of Hematology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai, China
| | - Fanli Hua
- Department of Hematology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai, China
| | - Hao Chen
- Department of Thoracic Surgery, Zhongshan Hospital Xuhui Branch, Fudan University, Shanghai, China
| | - Yunfeng Cheng
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Hematology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai, China.,Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.,Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Subotički T, Mitrović Ajtić O, Živković E, Diklić M, Đikić D, Tošić M, Beleslin-Čokić B, Dragojević T, Gotić M, Santibanez JF, Čokić V. VEGF Regulation of Angiogenic Factors via Inflammatory Signaling in Myeloproliferative Neoplasms. Int J Mol Sci 2021; 22:ijms22136671. [PMID: 34206393 PMCID: PMC8268588 DOI: 10.3390/ijms22136671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 01/10/2023] Open
Abstract
Background: Chronic inflammation has been recognized in neoplastic disorders, including myeloproliferative neoplasm (MPN), as an important regulator of angiogenesis. Aims: We investigated the influence of vascular endothelial growth factor (VEGF) and pro-inflammatory interleukin-6 (IL-6) on the expression of angiogenic factors, as well as inflammation-related signaling in mononuclear cells (MNC) of patients with MPN and JAK2V617F positive human erythroleukemic (HEL) cells. Results: We found that IL-6 did not change the expression of angiogenic factors in the MNC of patients with MPN and HEL cells. However, IL-6 and the JAK1/2 inhibitor Ruxolitinib significantly increased angiogenic factors—endothelial nitric oxide synthase (eNOS), VEGF, and hypoxia-inducible factor-1 alpha (HIF-1α)—in patients with polycythemia vera (PV). Furthermore, VEGF significantly increased the expression of HIF-1α and eNOS genes, the latter inversely regulated by PI3K and mTOR signaling in the MNC of primary myelofibrosis (PMF). VEGF and inhibitors of inflammatory JAK1/2, PI3K, and mTOR signaling reduced the eNOS protein expression in HEL cells. VEGF also decreased the expression of eNOS and HIF-1α proteins in the MNC of PMF. In contrast, VEGF increased eNOS and HIF-1α protein expression in the MNC of patients with PV, which was mediated by the inflammatory signaling. VEGF increased the level of IL-6 immunopositive MNC of MPN. In summary, VEGF conversely regulated gene and protein expression of angiogenic factors in the MNC of PMF, while VEGF increased angiogenic factor expression in PV mediated by the inflammation-related signaling. Conclusion: The angiogenic VEGF induction of IL-6 supports chronic inflammation that, through positive feedback, further promotes angiogenesis with concomitant JAK1/2 inhibition.
Collapse
Affiliation(s)
- Tijana Subotički
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (O.M.A.); (E.Ž.); (M.D.); (D.Đ.); (M.T.); (T.D.); (J.F.S.); (V.Č.)
- Correspondence: ; Tel.: +381-112685788
| | - Olivera Mitrović Ajtić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (O.M.A.); (E.Ž.); (M.D.); (D.Đ.); (M.T.); (T.D.); (J.F.S.); (V.Č.)
| | - Emilija Živković
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (O.M.A.); (E.Ž.); (M.D.); (D.Đ.); (M.T.); (T.D.); (J.F.S.); (V.Č.)
| | - Miloš Diklić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (O.M.A.); (E.Ž.); (M.D.); (D.Đ.); (M.T.); (T.D.); (J.F.S.); (V.Č.)
| | - Dragoslava Đikić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (O.M.A.); (E.Ž.); (M.D.); (D.Đ.); (M.T.); (T.D.); (J.F.S.); (V.Č.)
| | - Milica Tošić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (O.M.A.); (E.Ž.); (M.D.); (D.Đ.); (M.T.); (T.D.); (J.F.S.); (V.Č.)
| | - Bojana Beleslin-Čokić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Genetic Laboratory, Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | - Teodora Dragojević
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (O.M.A.); (E.Ž.); (M.D.); (D.Đ.); (M.T.); (T.D.); (J.F.S.); (V.Č.)
| | - Mirjana Gotić
- Clinic of Hematology, Clinical Center of Serbia, 11000 Belgrade, Serbia;
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Juan F. Santibanez
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (O.M.A.); (E.Ž.); (M.D.); (D.Đ.); (M.T.); (T.D.); (J.F.S.); (V.Č.)
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O’Higgins, Santiago 8370993, Chile
| | - Vladan Čokić
- Department of Molecular Oncology, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (O.M.A.); (E.Ž.); (M.D.); (D.Đ.); (M.T.); (T.D.); (J.F.S.); (V.Č.)
| |
Collapse
|
43
|
Christensen SF, Scherber RM, Mazza GL, Dueck AC, Brochmann N, Andersen CL, Hasselbalch HC, Mesa RA, Geyer HL. Tobacco use in the Myeloproliferative neoplasms: symptom burden, patient opinions, and care. BMC Cancer 2021; 21:691. [PMID: 34112113 PMCID: PMC8194237 DOI: 10.1186/s12885-021-08439-7] [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: 12/15/2020] [Accepted: 06/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients with Philadelphia-negative Myeloproliferative Neoplasms (MPN) suffer from numerous symptoms and decreased quality of life. Smoking is associated with an increased symptom burden in several malignancies. The aim of this study was to analyze the association between smoking and MPN-related symptom burden and explore MPN patients' opinions on smoking. METHODS A total of 435 patients with MPN participated in a cross-sectional internet-based survey developed by the Mayo Clinic and the Myeloproliferative Neoplasm Quality of Life Group. Patients reported their demographics, disease characteristics, tobacco use, and opinions on tobacco use. In addition, MPN-related symptoms were reported via the validated 10-item version of the Myeloproliferative Neoplasms Symptom Assessment Form. RESULTS Current/former smokers reported worse fatigue (mean severity 5.6 vs. 5.0, p = 0.02) and inactivity (mean severity 4.0 vs. 3.4, p = 0.03) than never smokers. Moreover, current/former smokers more frequently experienced early satiety (68.5% vs. 58.3%, p = 0.03), inactivity (79.9% vs. 71.1%, p = 0.04), and concentration difficulties (82.1% vs. 73.1%, p = 0.04). Although not significant, a higher total symptom burden was observed for current/former smokers (mean 30.4 vs. 27.0, p = 0.07). Accordingly, overall quality of life was significantly better among never smokers than current/former smokers (mean 3.5 vs. 3.9, p = 0.03). Only 43.2% of the current/former smokers reported having discussed tobacco use with their physician, and 17.5% did not believe smoking increased the risk of thrombosis. CONCLUSION The current study suggests that smoking may be associated with increased prevalence and severity of MPN symptoms and underscores the need to enhance patient education and address tobacco use in the care of MPN patients.
Collapse
Affiliation(s)
- Sarah F. Christensen
- Department of Hematology, Zealand University Hospital, Roskilde, Vestermarksvej 9, 4000 Roskilde, Denmark
| | - Robyn M. Scherber
- Department of Hematology and Oncology, UT Health San Antonio MD Anderson Cancer Center, 7979 Wurzbach Rd, San Antonio, TX 78229 USA
- Hematologic Malignancies, Incyte Corporation, Wilmington, Delaware, USA
| | - Gina L. Mazza
- Department of Health Sciences Research, Mayo Clinic, 13400 East Shea Boulevard, Scottsdale, AZ 85259 USA
| | - Amylou C. Dueck
- Department of Health Sciences Research, Mayo Clinic, 13400 East Shea Boulevard, Scottsdale, AZ 85259 USA
| | - Nana Brochmann
- Department of Hematology, Zealand University Hospital, Roskilde, Vestermarksvej 9, 4000 Roskilde, Denmark
| | - Christen L. Andersen
- Department of Hematology, University Hospital of Copenhagen at Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Hans C. Hasselbalch
- Department of Hematology, Zealand University Hospital, Roskilde, Vestermarksvej 9, 4000 Roskilde, Denmark
| | - Ruben A. Mesa
- Department of Hematology and Oncology, UT Health San Antonio MD Anderson Cancer Center, 7979 Wurzbach Rd, San Antonio, TX 78229 USA
| | - Holly L. Geyer
- Department of Hematology and Medical Oncology, Mayo Clinic, 13400 East Shea Boulevard, Scottsdale, AZ 85259 USA
| |
Collapse
|
44
|
Fisher DAC, Fowles JS, Zhou A, Oh ST. Inflammatory Pathophysiology as a Contributor to Myeloproliferative Neoplasms. Front Immunol 2021; 12:683401. [PMID: 34140953 PMCID: PMC8204249 DOI: 10.3389/fimmu.2021.683401] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Myeloid neoplasms, including acute myeloid leukemia (AML), myeloproliferative neoplasms (MPNs), and myelodysplastic syndromes (MDS), feature clonal dominance and remodeling of the bone marrow niche in a manner that promotes malignant over non-malignant hematopoiesis. This take-over of hematopoiesis by the malignant clone is hypothesized to include hyperactivation of inflammatory signaling and overproduction of inflammatory cytokines. In the Ph-negative MPNs, inflammatory cytokines are considered to be responsible for a highly deleterious pathophysiologic process: the phenotypic transformation of polycythemia vera (PV) or essential thrombocythemia (ET) to secondary myelofibrosis (MF), and the equivalent emergence of primary myelofibrosis (PMF). Bone marrow fibrosis itself is thought to be mediated heavily by the cytokine TGF-β, and possibly other cytokines produced as a result of hyperactivated JAK2 kinase in the malignant clone. MF also features extramedullary hematopoiesis and progression to bone marrow failure, both of which may be mediated in part by responses to cytokines. In MF, elevated levels of individual cytokines in plasma are adverse prognostic indicators: elevated IL-8/CXCL8, in particular, predicts risk of transformation of MF to secondary AML (sAML). Tumor necrosis factor (TNF, also known as TNFα), may underlie malignant clonal dominance, based on results from mouse models. Human PV and ET, as well as MF, harbor overproduction of multiple cytokines, above what is observed in normal aging, which can lead to cellular signaling abnormalities separate from those directly mediated by hyperactivated JAK2 or MPL kinases. Evidence that NFκB pathway signaling is frequently hyperactivated in a pan-hematopoietic pattern in MPNs, including in cells outside the malignant clone, emphasizes that MPNs are pan-hematopoietic diseases, which remodel the bone marrow milieu to favor persistence of the malignancy. Clinical evidence that JAK2 inhibition by ruxolitinib in MF neither reliably reduces malignant clonal burden nor eliminates cytokine elevations, suggests targeting cytokine mediated signaling as a therapeutic strategy, which is being pursued in new clinical trials. Greater knowledge of inflammatory pathophysiology in MPNs can therefore contribute to the development of more effective therapy.
Collapse
Affiliation(s)
- Daniel Arthur Corpuz Fisher
- Divisions of Hematology & Oncology, School of Medicine, Washington University in St. Louis, Saint Louis, MO, United States
| | - Jared Scott Fowles
- Divisions of Hematology & Oncology, School of Medicine, Washington University in St. Louis, Saint Louis, MO, United States
| | - Amy Zhou
- Divisions of Hematology & Oncology, School of Medicine, Washington University in St. Louis, Saint Louis, MO, United States
| | - Stephen Tracy Oh
- Divisions of Hematology & Oncology, School of Medicine, Washington University in St. Louis, Saint Louis, MO, United States
| |
Collapse
|
45
|
Rolfes V, Ribeiro LS, Hawwari I, Böttcher L, Rosero N, Maasewerd S, Santos MLS, Próchnicki T, Silva CMDS, Wanderley CWDS, Rothe M, Schmidt SV, Stunden HJ, Bertheloot D, Rivas MN, Fontes CJ, Carvalho LH, Cunha FQ, Latz E, Arditi M, Franklin BS. Platelets Fuel the Inflammasome Activation of Innate Immune Cells. Cell Rep 2021; 31:107615. [PMID: 32402278 PMCID: PMC7225754 DOI: 10.1016/j.celrep.2020.107615] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/12/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The inflammasomes control the bioactivity of pro-inflammatory cytokines of the interleukin (IL)-1 family. The inflammasome assembled by NLRP3 has been predominantly studied in homogeneous cell populations in vitro, neglecting the influence of cellular interactions that occur in vivo. Here, we show that platelets boost the inflammasome capacity of human macrophages and neutrophils and are critical for IL-1 production by monocytes. Platelets license NLRP3 transcription, thereby enhancing ASC oligomerization, caspase-1 activity, and IL-1β secretion. Platelets influence IL-1β production in vivo, and blood platelet counts correlate with plasmatic IL-1β levels in malaria. Furthermore, we reveal an enriched platelet gene signature among the highest-expressed transcripts in IL-1β-driven autoinflammatory diseases. The platelet effect is independent of cell-to-cell contact, platelet-derived lipid mediators, purines, nucleic acids, and a host of platelet cytokines, and it involves the triggering of calcium-sensing receptors on macrophages. Hence, platelets provide an additional layer of regulation of inflammasomes and IL-1-driven inflammation. Platelets license NLRP3 for inflammasome activattion in innate immune cells Platelets are required for optimal monocyte inflammasome activation Platelets shape IL-1β in vivo, and platelet counts correlate with IL-1β in plasma A constitutive, heat-sensitive soluble platelet-factor boost IL-1β in macrophages
Collapse
Affiliation(s)
- Verena Rolfes
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Lucas Secchim Ribeiro
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany.
| | - Ibrahim Hawwari
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Lisa Böttcher
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Nathalia Rosero
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Salie Maasewerd
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Marina Lima Silva Santos
- Laboratório de Malária, Instituto René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, MG, Brazil
| | - Tomasz Próchnicki
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Camila Meirelles de Souza Silva
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Carlos Wagner de Souza Wanderley
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Maximilian Rothe
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Susanne V Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - H James Stunden
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Damien Bertheloot
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Magali Noval Rivas
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Cor Jesus Fontes
- Departamento de Clínica Médica, Universidade Federal de Mato Grosso, 78060-900 Cuiabá, MT, Brazil
| | - Luzia Helena Carvalho
- Laboratório de Malária, Instituto René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, MG, Brazil
| | - Fernando Queiroz Cunha
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Eicke Latz
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01655, USA; German Center for Neurodegenerative Diseases, 53127 Bonn, NRW, Germany
| | - Moshe Arditi
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA.
| | | |
Collapse
|
46
|
Cominal JG, Cacemiro MDC, Berzoti-Coelho MG, Pereira IEG, Frantz FG, Souto EX, Covas DT, de Figueiredo-Pontes LL, Oliveira MC, Malmegrim KCR, de Castro FA. Bone Marrow Soluble Mediator Signatures of Patients With Philadelphia Chromosome-Negative Myeloproliferative Neoplasms. Front Oncol 2021; 11:665037. [PMID: 34084749 PMCID: PMC8167065 DOI: 10.3389/fonc.2021.665037] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
Background Essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF) are clonal hematological diseases classified as Philadelphia chromosome-negative myeloproliferative neoplasms (MPN). MPN pathogenesis is associated with the presence of somatic driver mutations, bone marrow (BM) niche alterations, and tumor inflammatory status. The relevance of soluble mediators in the pathogenesis of MPN led us to analyze the levels of cytokines, chemokines, and growth factors related to inflammation, angiogenesis and hematopoiesis regulation in the BM niche of MPN patients. Methods Soluble mediator levels in BM plasma samples from 17 healthy subjects, 28 ET, 19 PV, and 16 PMF patients were determined using a multiplex assay. Soluble mediator signatures were created from categorical analyses of high mediator producers. Soluble mediator connections and the correlation between plasma levels and clinic-laboratory parameters were also analyzed. Results The soluble mediator signatures of the BM niche of PV patients revealed a highly inflammatory and pro-angiogenic milieu, with increased levels of chemokines (CCL2, CCL5, CXCL8, CXCL12, CXCL10), and growth factors (GM-CSF M-CSF, HGF, IFN-γ, IL-1β, IL-6Ra, IL-12, IL-17, IL-18, TNF-α, VEGF, and VEGF-R2). ET and PMF patients presented intermediate inflammatory and pro-angiogenic profiles. Deregulation of soluble mediators was associated with some clinic-laboratory parameters of MPN patients, including vascular events, treatment status, risk stratification of disease, hemoglobin concentration, hematocrit, and red blood cell count. Conclusions Each MPN subtype exhibits a distinct soluble mediator signature. Deregulated production of BM soluble mediators may contribute to MPN pathogenesis and BM niche modification, provides pro-tumor stimuli, and is a potential target for future therapies.
Collapse
Affiliation(s)
- Juçara Gastaldi Cominal
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maira da Costa Cacemiro
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria Gabriela Berzoti-Coelho
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Illy Enne Gomes Pereira
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fabiani Gai Frantz
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Elizabeth Xisto Souto
- Department of Clinical Hematology, Euryclides de Jesus Zerbini Transplant Hospital, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Lorena Lobo de Figueiredo-Pontes
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Division of Hematology, Hemotherapy and Cellular Therapy, Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria Carolina Oliveira
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Division of Rheumatology, Allergy and Immunotherapy, Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kelen Cristina Ribeiro Malmegrim
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fabíola Attié de Castro
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
47
|
Jain A, Deo P, Sachdeva MUS, Bose P, Lad D, Prakash G, Khadwal A, Varma N, Varma S, Malhotra P. Aberrant expression of cytokines in polycythemia vera correlate with the risk of thrombosis. Blood Cells Mol Dis 2021; 89:102565. [PMID: 33831662 DOI: 10.1016/j.bcmd.2021.102565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Thrombo-hemorrhagic complications cause significant morbidity and mortality in patients with polycythemia vera (PV). OBJECTIVES To assay and correlate inflammatory cytokines with the thrombotic risk in PV patients. METHODS This prospective observational study was carried out at Postgraduate Institute of Medical Education and Research, Chandigarh, India over 18-months. The study enrolled 52 patients with PV (newly diagnosed = 28, follow-up = 24), and 20 age/sex-matched controls. Cytokine analysis for IL 1β, IL2, IL4, IL6, IL8, IL10, IL11, IL12/23p40, TNFα, and IFN-γ was performed on the peripheral blood (before treatment initiation for newly diagnosed cases, and after 7 days of stopping drugs for follow-up cases) by flow cytometry-based cytokine bead analysis (CBA) using CBA kits (BD™ biosciences, USA). Results were analyzed using SPSS Statistics 22.0. RESULTS The mean age of patients was 51.9 ± 13 years. Levels of IL-6, IL-1β, IL-8, IL-11, IL-12/23p40 were significantly raised, however, TNF-α, and IFN-γ levels were significantly lower in the PV population as compared to controls. A significant correlation between the levels of IL-6, IL-2, and IL-8 with the overall risk of thrombosis in PV patients was observed. CONCLUSIONS PV patients display an aberrant pattern of plasma cytokine expression, the levels of which correlate with the thrombotic risk.
Collapse
Affiliation(s)
- Ankur Jain
- Department of Haematology, Vardhman Mahavir Medical College and Safdarjung Hospital, Delhi 110029, India
| | - Prateek Deo
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Man Updesh Singh Sachdeva
- Department of Haematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Parveen Bose
- Department of Haematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepesh Lad
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gaurav Prakash
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Khadwal
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Neelam Varma
- Department of Haematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Subhash Varma
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Malhotra
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
48
|
Mesa R, Palmer J, Eckert R, Huberty J. Quality of Life in Myeloproliferative Neoplasms: Symptoms and Management Implications. Hematol Oncol Clin North Am 2021; 35:375-390. [PMID: 33641875 DOI: 10.1016/j.hoc.2020.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Myeloproliferative neoplasms include essential thrombocythemia, polycythemia vera, and myelofibrosis. They are characterized by abnormal myeloid proliferation. Patients suffer from debilitating constitutional symptoms and splenomegaly. There have been advances in understanding the impact on quality of life in myeloproliferative neoplasms. Owing to the chronicity of these diseases, symptoms are considered in response criteria for clinical trials. This review wills cover how quality of life is measured in patients with myeloproliferative neoplasm. We review the impact of treatment options, including JAK inhibitors, allogeneic stem cell transplantation, and medications in development. We discuss nonpharmacologic methods of improving symptoms and quality of life.
Collapse
Affiliation(s)
- Ruben Mesa
- Mays Cancer Center, UT Health San Antonio MD Anderson, 7979 Wurzbach Road, San Antonio, TX 78229, USA.
| | - Jeanne Palmer
- Division of Hematology & Medical Oncology, Mayo Clinic, 5777 East Mayo boulevard, Phoenix, AZ, 85054, USA
| | - Ryan Eckert
- Mays Cancer Center, UT Health San Antonio MD Anderson, 7979 Wurzbach Road, San Antonio, TX 78229, USA; Arizona State University College of Health Solutions, 550 North 3rd Street, Phoenix, AZ 85004, USA
| | - Jennifer Huberty
- Mays Cancer Center, UT Health San Antonio MD Anderson, 7979 Wurzbach Road, San Antonio, TX 78229, USA; Arizona State University College of Health Solutions, 550 North 3rd Street, Phoenix, AZ 85004, USA
| |
Collapse
|
49
|
Guijarro-Hernández A, Vizmanos JL. A Broad Overview of Signaling in Ph-Negative Classic Myeloproliferative Neoplasms. Cancers (Basel) 2021; 13:cancers13050984. [PMID: 33652860 PMCID: PMC7956519 DOI: 10.3390/cancers13050984] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary There is growing evidence that Ph-negative myeloproliferative neoplasms are disorders in which multiple signaling pathways are significantly disturbed. The heterogeneous phenotypes observed among patients have highlighted the importance of having a comprehensive knowledge of the molecular mechanisms behind these diseases. This review aims to show a broad overview of the signaling involved in myeloproliferative neoplasms (MPNs) and other processes that can modify them, which could be helpful to better understand these diseases and develop more effective targeted treatments. Abstract Ph-negative myeloproliferative neoplasms (polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF)) are infrequent blood cancers characterized by signaling aberrations. Shortly after the discovery of the somatic mutations in JAK2, MPL, and CALR that cause these diseases, researchers extensively studied the aberrant functions of their mutant products. In all three cases, the main pathogenic mechanism appears to be the constitutive activation of JAK2/STAT signaling and JAK2-related pathways (MAPK/ERK, PI3K/AKT). However, some other non-canonical aberrant mechanisms derived from mutant JAK2 and CALR have also been described. Moreover, additional somatic mutations have been identified in other genes that affect epigenetic regulation, tumor suppression, transcription regulation, splicing and other signaling pathways, leading to the modification of some disease features and adding a layer of complexity to their molecular pathogenesis. All of these factors have highlighted the wide variety of cellular processes and pathways involved in the pathogenesis of MPNs. This review presents an overview of the complex signaling behind these diseases which could explain, at least in part, their phenotypic heterogeneity.
Collapse
Affiliation(s)
- Ana Guijarro-Hernández
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
| | - José Luis Vizmanos
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Correspondence:
| |
Collapse
|
50
|
Nasillo V, Riva G, Paolini A, Forghieri F, Roncati L, Lusenti B, Maccaferri M, Messerotti A, Pioli V, Gilioli A, Bettelli F, Giusti D, Barozzi P, Lagreca I, Maffei R, Marasca R, Potenza L, Comoli P, Manfredini R, Maiorana A, Tagliafico E, Luppi M, Trenti T. Inflammatory Microenvironment and Specific T Cells in Myeloproliferative Neoplasms: Immunopathogenesis and Novel Immunotherapies. Int J Mol Sci 2021; 22:ijms22041906. [PMID: 33672997 PMCID: PMC7918142 DOI: 10.3390/ijms22041906] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
The Philadelphia-negative myeloproliferative neoplasms (MPNs) are malignancies of the hematopoietic stem cell (HSC) arising as a consequence of clonal proliferation driven by somatically acquired driver mutations in discrete genes (JAK2, CALR, MPL). In recent years, along with the advances in molecular characterization, the role of immune dysregulation has been achieving increasing relevance in the pathogenesis and evolution of MPNs. In particular, a growing number of studies have shown that MPNs are often associated with detrimental cytokine milieu, expansion of the monocyte/macrophage compartment and myeloid-derived suppressor cells, as well as altered functions of T cells, dendritic cells and NK cells. Moreover, akin to solid tumors and other hematological malignancies, MPNs are able to evade T cell immune surveillance by engaging the PD-1/PD-L1 axis, whose pharmacological blockade with checkpoint inhibitors can successfully restore effective antitumor responses. A further interesting cue is provided by the recent discovery of the high immunogenic potential of JAK2V617F and CALR exon 9 mutations, that could be harnessed as intriguing targets for innovative adoptive immunotherapies. This review focuses on the recent insights in the immunological dysfunctions contributing to the pathogenesis of MPNs and outlines the potential impact of related immunotherapeutic approaches.
Collapse
Affiliation(s)
- Vincenzo Nasillo
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy; (G.R.); (B.L.); (E.T.); (T.T.)
- Correspondence: ; Tel.: +39-059-422-2173
| | - Giovanni Riva
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy; (G.R.); (B.L.); (E.T.); (T.T.)
| | - Ambra Paolini
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Fabio Forghieri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Luca Roncati
- Institute of Pathology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (L.R.); (A.M.)
| | - Beatrice Lusenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy; (G.R.); (B.L.); (E.T.); (T.T.)
| | - Monica Maccaferri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Andrea Messerotti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Valeria Pioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Andrea Gilioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Francesca Bettelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Davide Giusti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Patrizia Barozzi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Ivana Lagreca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Rossana Maffei
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Roberto Marasca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Leonardo Potenza
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Patrizia Comoli
- Pediatric Hematology/Oncology Unit and Cell Factory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, 27100 Pavia, Italy;
| | - Rossella Manfredini
- Centre for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Antonino Maiorana
- Institute of Pathology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (L.R.); (A.M.)
| | - Enrico Tagliafico
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy; (G.R.); (B.L.); (E.T.); (T.T.)
| | - Mario Luppi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico, 41124 Modena, Italy; (A.P.); (F.F.); (M.M.); (A.M.); (V.P.); (A.G.); (F.B.); (D.G.); (P.B.); (I.L.); (R.M.); (R.M.); (L.P.); (M.L.)
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy; (G.R.); (B.L.); (E.T.); (T.T.)
| |
Collapse
|