1
|
Leiva O, Soo S, Smilowitz NR, Reynolds H, Shah B, Bernard S, How J, Lee MH, Hobbs G. Impact of Echocardiographic Probability of Pulmonary Hypertension on Prognosis and Outcomes Among Patients With Myeloproliferative Neoplasms. Circ Cardiovasc Imaging 2025:e017986. [PMID: 40492300 DOI: 10.1161/circimaging.124.017986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 05/19/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND Myeloproliferative neoplasms (MPN) are a group of chronic leukemias that are associated with pulmonary hypertension (PH), which has been associated with increased risk adverse outcomes. The echocardiographic characterization of PH in MPN has not been reported, and the prognostic significance of PH among patients with MPN remains unclear. METHODS Multicenter, retrospective cohort study of patients with MPN with ≥1 echocardiogram from 2010 to 2023. The echocardiographic probability of PH was determined according to the guidelines. The outcomes were hematologic progression and major adverse cardiovascular events. Exploratory analysis included outcomes among patients with right heart catheterization after the first echocardiogram, with PH defined as mean pulmonary artery pressure of >20 mm Hg. Multivariable Fine-Gray competing risk regression was used to estimate the subhazard ratio of hematologic progression and major adverse cardiovascular events. RESULTS Five hundred fifty-five patients were included and 237 (42.7%) had an intermediate or high probability of PH on echocardiography. Over a median follow-up period of 51.2 months (interquartile range, 29.5-79.8), it was observed that echocardiographic probability of PH was associated with increased risk of hematologic progression (adjusted subhazard ratio, 1.92 [95% CI, 1.09-3.39]) and major adverse cardiovascular events (adjusted subhazard ratio, 1.66 [95% CI, 1.04-2.66]) but not all-cause death (adjusted hazard ratio, 1.51 [95% CI, 0.98-2.32]). Among patients with right heart catheterization (n=61), PH was present in 51 (83.6%) of patients and was associated with a higher risk of hematologic progression (29.4% versus 0%; P=0.048). CONCLUSIONS Among patients with MPN, echocardiographic probability of PH was associated with an increased risk of hematologic progression. Prospective studies are needed to assess the optimal use of echocardiography on MPN-specific prognostication.
Collapse
Affiliation(s)
- Orly Leiva
- Section of Cardiology-Heart Failure, Department of Medicine, University of Chicago, IL (O.L.)
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (O.L., N.R.S., H.R., B.S., S.B.)
| | - Steven Soo
- Department of Medicine, New York University Long Island School of Medicine, Mineola (S.S.)
| | - Nathaniel R Smilowitz
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (O.L., N.R.S., H.R., B.S., S.B.)
| | - Harmony Reynolds
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (O.L., N.R.S., H.R., B.S., S.B.)
| | - Binita Shah
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (O.L., N.R.S., H.R., B.S., S.B.)
| | - Samuel Bernard
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (O.L., N.R.S., H.R., B.S., S.B.)
| | - Joan How
- Division of Hematology and Oncology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (J.H.)
| | - Michelle Hyunju Lee
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (M.H.L., G.H.)
| | - Gabriela Hobbs
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston (M.H.L., G.H.)
| |
Collapse
|
2
|
Zhang N, Tian X, Sun D, Tse G, Xie B, Zhao Z, Liu T. Clonal hematopoiesis, cardiovascular disease and cancer treatment-induced cardiotoxicity. Semin Cancer Biol 2025; 111:89-114. [PMID: 40023267 DOI: 10.1016/j.semcancer.2025.02.007] [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: 04/23/2024] [Revised: 01/05/2025] [Accepted: 02/06/2025] [Indexed: 03/04/2025]
Abstract
Clonal hematopoiesis (CH) arises when a substantial proportion of mature blood cells is derived from a single hematopoietic stem cell lineage. It is considered to be a premalignant state that predisposes individuals to an increased risk of cancers. Recently, emerging evidence has demonstrated a strong association between CH and both the incidence and mortality of cardiovascular diseases (CVD), with the relative risks being comparable to those attributed to traditional cardiovascular risk factors. In addition, CH has been suggested to play a role in CVD and anti-cancer treatment-related cardiotoxicity amongst cancer survivors. Moreover, certain forms of chemotherapy and radiation therapy have been shown to promote the clonal expansion of specific CH-related mutations. Consequently, CH may play a substantial role in the realm of cardio-oncology. In this review, we discuss the association between CH with cancer and CVD, with a special focus on anti-cancer treatment-related cardiotoxicity, discuss possible future research avenues and propose a systematic approach for clinical practice.
Collapse
Affiliation(s)
- Nan Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xu Tian
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Dongkun Sun
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China; School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| | - Bingxin Xie
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
3
|
Redondo S. Myeloproliferative neoplasms: A model of the journey from clonal hematopoiesis to cardiovascular disease and cancer. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2025:500767. [PMID: 40413094 DOI: 10.1016/j.arteri.2025.500767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 05/27/2025]
Abstract
In the last decade, the coming of next-generation sequencing and its application to large human populations is breaking the barrier between inflammation and cancer. Indeed, acquired mutations in key genes that regulate hematopoiesis and thus confer a selective advantage in the proliferation of hematopoietic progenitors have established the concept of clonal hematopoiesis of indeterminate potential or CHIP. A growing body of clinical and experimental evidence is highlighting the link between CHIP and adverse outcomes, in particular atherosclerotic cardiovascular disease and cancer. The apparent surprise about how these two different entities share common mechanisms can be explained by myeloproliferation and inflammation. These mechanisms are involved not only in the development of myeloid tumors but also in atherogenesis. Myeloproliferative neoplasms or MPN are a type of myeloid tumors where thrombotic risk is increased not only by higher blood counts but also by means of an accelerated atherosclerosis. Therefore, myeloproliferative neoplasms are a model of the link between clonal hematopoiesis and atherosclerotic cardiovascular disease. The concept of CHIP has important clinical applications. A deeper understanding of these mechanisms may pave the way for the future early diagnosis and potential pre-emptive treatments of these two major causes of death.
Collapse
|
4
|
Yoon SY, Kim M, Kim H, Bang DW, Park BW, Jeong SY, Lee MY, Kim KH, Lee N, Won JH, Moon I, Suh J, Kwon SS. Risk of Hematologic Malignancies in Patients With Acute Myocardial Infarction: A Nationwide Population-Based Cohort Study. JACC CardioOncol 2025:S2666-0873(25)00146-2. [PMID: 40434327 DOI: 10.1016/j.jaccao.2025.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/01/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) and cancer are leading causes of death worldwide. However, the relationship between AMI and hematologic malignancies remains unclear. OBJECTIVES The authors aimed to investigate the association between AMI and the subsequent risk of incident hematologic malignancies. METHODS This retrospective cohort study included 103,686 patients with AMI and no history of hematologic malignancies, and 103,686 age- and sex-matched individuals with no history of AMI or hematologic malignancies, diagnosed between January 1, 2003, and December 31, 2021. Data were obtained from the Korean National Health Insurance claims database. We compared the cumulative incidence of hematologic malignancies between groups using Gray's method. HRs and 95% CIs were calculated using Gray's competing risk regression model, with death treated as a competing risk. RESULTS During follow-up (AMI, 7.9 years [Q1-Q3: 5.2-11.4 years]; control group, 17.8 years [Q1-Q3: 14.8-17.9 years]), 1,043 and 1,479 individuals in the AMI and control groups, respectively, were newly diagnosed with hematologic malignancies (incidence rate per 1,000 person-years: 1.21 vs 0.93). Competing risk analysis revealed that the AMI group had a higher risk of hematologic malignancy than the control group (HR: 1.49; 95% CI: 1.31-1.69). Findings were consistent in sensitivity and standardized incidence ratio analyses. CONCLUSIONS Patients with AMI had a higher risk of hematologic malignancies than those without AMI. These findings suggest an association between AMI and hematologic malignancies, and underscore the importance of considering hematologic malignancy development in patients with AMI.
Collapse
Affiliation(s)
- Seug Yun Yoon
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Mina Kim
- Department of Data Science, Hanmi Pharm. Co., Ltd, Seoul, Republic of Korea
| | - Hoseob Kim
- Department of Data Science, Hanmi Pharm. Co., Ltd, Seoul, Republic of Korea
| | - Duk Won Bang
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Byoung-Won Park
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Sun Young Jeong
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Min-Young Lee
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Kyoung Ha Kim
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Namsu Lee
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Jong-Ho Won
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Inki Moon
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Jon Suh
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Seong Soon Kwon
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Shimosato H, Usui E, Sayama K, Kanaji Y, Hada M, Nagamine T, Ueno H, Setoguchi M, Watanabe T, Kakuta T. Characteristic Findings of Honeycomb-Like Structures in Patients With Chronic Coronary Syndrome Complicated With Myeloproliferative Neoplasms. JACC Case Rep 2025; 30:103680. [PMID: 40185592 PMCID: PMC12046759 DOI: 10.1016/j.jaccas.2025.103680] [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: 11/07/2024] [Accepted: 12/18/2024] [Indexed: 04/07/2025]
Abstract
A 76-year-old woman with a history of essential thrombocythemia and a 64-year-old man with a history of polycythemia vera were both found to have coronary artery stenosis on coronary computed tomographic angiography. Intravascular imaging revealed a honeycomb-like structure with no lipid component, suggesting an organizing thrombus associated with myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Hikaru Shimosato
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tokyo, Japan
| | - Eisuke Usui
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tokyo, Japan
| | - Kodai Sayama
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tokyo, Japan
| | - Yoshihisa Kanaji
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tokyo, Japan
| | - Masahiro Hada
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tokyo, Japan
| | - Tatsuhiro Nagamine
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tokyo, Japan
| | - Hiroki Ueno
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tokyo, Japan
| | - Mirei Setoguchi
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tokyo, Japan
| | - Takahiro Watanabe
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tokyo, Japan
| | - Tsunekazu Kakuta
- Department of Cardiovascular Medicine, Tsuchiura Kyodo General Hospital, Tokyo, Japan.
| |
Collapse
|
6
|
Leiva O, Lee MH, How J, Berger JS, Hobbs G. Outcomes of patients with myeloproliferative neoplasms and critical limb ischemia: insights from the National readmissions database. J Thromb Thrombolysis 2025:10.1007/s11239-025-03088-0. [PMID: 40131642 DOI: 10.1007/s11239-025-03088-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
Patients with myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF), are at increased risk of atherosclerosis, including peripheral arterial disease (PAD). Critical limb ischemia (CLI) may complicate PAD and is associated with significant mortality and morbidity. Despite the increased risk of thrombosis with MPN, outcomes of CLI in MPN patients are unclear. We conducted an analysis utilizing the 2017-2020 National Readmission Database (NRD) of patients hospitalized for CLI with and without MPN. Patients with MPN were propensity score matched (PSM) with patients without MPN. Primary outcome was composite outcome of major adverse cardiovascular and limb events (MACLE). Logistic regression was utilized to estimate risk of MACLE in patients with MPN vs. without MPN. Inverse-probability treatment weighted (IPTW) analysis was performed to evaluate the effect of revascularization on MACLE in patients with MPN. A total of 102,598 patients were included, 931 (0.9%) had MPN. After PSM, MPN was associated with increased risk of MACLE (47.3% vs. 39.1%; OR 1.40, 95% CI 1.21-1.62). After IPTW, revascularization was associated with decreased risk of MACLE among patients with MPN (45.0% vs. 50.7%; OR 0.80, 95% CI 0.66-0.96). Among patients admitted with CLI, MPN was associated with increased risk of MACLE especially ET and MF phenotypes. Revascularization was associated with decreased risk of MACLE among patients with MPN. Further investigation is needed in order to improve outcomes in patients with MPN and CLI.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
- Section of Cardiology- Heart Failure, Department of Medicine, University of Chicago, Chicago, IL, USA.
| | - Michelle H Lee
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joan How
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeffrey S Berger
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Gabriela Hobbs
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Ang SP, Chia JE, Krittanawong C, Piana RN, Lee K, Ayoub C, Pineda JE, Song D, Mukherjee D. Trends and Outcomes Following Percutaneous Coronary Intervention in Patients With Myeloproliferative Neoplasms: Insights From National Database. Catheter Cardiovasc Interv 2025. [PMID: 40079618 DOI: 10.1002/ccd.31489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/22/2025] [Accepted: 03/01/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Myeloproliferative neoplasms (MPN) are associated with an increased cardiovascular risk including acute coronary syndrome. However, there is a lack of comprehensive data regarding the rate of percutaneous coronary intervention (PCI), as well as the in-hospital characteristics and outcomes for MPN patients. AIMS We aimed to evaluate the temporal trends and outcomes of PCI among patients with MPN. METHODS AND RESULTS The National Inpatient Sample database from 2016 to 2020 was queried to identify all PCI hospitalizations. Temporal trends and outcomes of patients with and without MPN following PCI were analyzed. Propensity score matching (PSM) was implemented to compare outcomes between MPN and non-MPN groups. 2,237,210 PCI hospitalizations with 7560 (0.27%) patients with MPN were included in this study. Throughout the study period, the prevalence of MPN among PCI admissions remained stable (p-value for trend = 0.12). Within the MPN subgroup, essential thrombocythemia (ET) was the predominant condition (53.2). Patients with MPN had higher prevalence of cardiovascular comorbidities than non-MPN patients. Following PSM, MPNs were significantly associated with a higher risk of blood transfusions (OR: 1.66, 95% CI: 1.22-2.24, p = 0.001) and AKI (OR: 1.39, 95% CI: 1.17-1.65, p < 0.001). In contrast, the risk of in-hospital mortality (OR: 1.18, 95% CI: 0.83-1.69, p = 0.354 and bleeding (OR: 1.43, 95% CI: 0.90-2.27, p = 0.127) did not significantly differ between the two groups. CONCLUSIONS Our study demonstrated that while the prevalence of MPN among patients undergoing PCI remained stable, those with MPN faced higher risks of bleeding, blood transfusion and acute kidney injury. Further research is warranted to explore the underlying reasons for these increased risks and to improve outcomes in this high-risk group.
Collapse
Affiliation(s)
- Song Peng Ang
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, New Jersey, USA
| | - Jia Ee Chia
- Department of Internal Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | | | - Robert N Piana
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kwan Lee
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Jr Exequiel Pineda
- Department of Cardiovascular Medicine, University of Arizona, Tucson, Arizona, USA
| | - David Song
- Department of Cardiovascular Medicine, New York Presbyterian, Queens, New York, USA
| | - Debabrata Mukherjee
- Department of Cardiovascular Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| |
Collapse
|
8
|
Li A, Kaphle R, Zanotti G, Chaudhry SP. Regression of Left Ventricular Hypertrophy After Polycythemia Vera Treatment With Incidental Finding of Unicuspid Aortic Valve. JACC Case Rep 2025; 30:103176. [PMID: 40054962 PMCID: PMC11911843 DOI: 10.1016/j.jaccas.2024.103176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/23/2024] [Accepted: 11/18/2024] [Indexed: 03/20/2025]
Abstract
We present a unique case of a 38-year-old man with 2 rare conditions: unicuspid aortic valve and polycythemia vera. The patient developed left ventricular hypertrophy, which showed regression after polycythemia vera treatment. This is the first reported case of left ventricular hypertrophy regression after polycythemia vera treatment.
Collapse
Affiliation(s)
- Anita Li
- Department of Internal Medicine, Ascension St. Vincent Hospital, Indianapolis, Indiana, USA.
| | - Roshan Kaphle
- Department of Internal Medicine, Ascension St. Vincent Hospital, Indianapolis, Indiana, USA
| | - Giorgio Zanotti
- Department of Cardiovascular Surgery, Ascension St. Vincent Hospital, Indianapolis, Indiana, USA
| | - Sunit-Preet Chaudhry
- Department of Cardiology, Ascension St. Vincent Hospital, Indianapolis, Indiana, USA
| |
Collapse
|
9
|
Leiva O, Zhou S, How J, Lee M, Hobbs G. Readmission outcomes after acute coronary syndrome among patients with myeloproliferative neoplasms. Atherosclerosis 2025; 401:119046. [PMID: 39700748 DOI: 10.1016/j.atherosclerosis.2024.119046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND AND AIMS Myeloproliferative neoplasms (MPNs) are associated with arterial thrombosis, including acute coronary syndrome (ACS). Prior studies have suggested similar in-hospital mortality among patients with MPN compared to those without. However, post-ACS outcomes have not been thoroughly evaluated. METHODS Patients hospitalized for ACS with and without MPN from January 2014 to December 2020 were identified using the National Readmission Database (NRD). Primary outcome was 90- and 180-day cardiovascular (CV) readmissions. Secondary outcomes were 90- and 180-day arterial thrombosis (AT), heart failure (HF), bleeding, and all-cause readmission and index hospitalization death, bleeding and arterial thrombosis (including ischemic stroke and arterial thromboembolism). Propensity score matching was used to compare outcomes between patients with and without MPN. RESULTS After PSM, 8667 patients with MPN were matched with 43,335 patients without MPN. MPN was associated with increased risk of 90- (HR 1.22, 95 % CI 1.13-1.32) and 180-day (HR 1.22, 95 % CI 1.12-1.32) readmissions. MPN was also associated with increased risk of 90- and 180-day AT, HF, bleeding, and all-cause readmissions. Among patients with MPN, MF was associated with increased risk of 90- (HR 1.36, 95 % CI 1.24-1.50) and 180- day (HR 1.34, 95 % CI 1.21-1.48) readmissions. CONCLUSIONS MPN was associated with increased risk of 90- and 180-day readmissions among patients hospitalized for ACS. Among patients with MPN, MF was associated with increased risk of 90- and 180-day CV readmissions. Further investigation is needed to improve post-ACS outcomes among patients with MPN.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA; Section of Cardiology - Heart Failure, Department of Medicine, University of Chicago, Chicago, IL, USA.
| | - Sophia Zhou
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Joan How
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, USA
| | - Michelle Lee
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gabriela Hobbs
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Manganas K, Bemplidakis T, Papachristou K, Angelara M, Karamanakos G. Multiple Aneurysms and Thrombotic Events as Initial Manifestations of Primary Myelofibrosis: A Case Report. Cureus 2025; 17:e79519. [PMID: 40135024 PMCID: PMC11936429 DOI: 10.7759/cureus.79519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2025] [Indexed: 03/27/2025] Open
Abstract
This case report presents a 66-year-old male who developed deep venous thrombosis (DVT), pulmonary embolism (PE), and a ruptured iliac aneurysm as initial manifestations of primary myelofibrosis (PMF). Due to the presence of pre-existing aneurysms in combination with anticoagulation therapy, the patient experienced a retroperitoneal hematoma, necessitating temporary cessation of treatment. Genetic testing revealed a JAK2 V617F mutation and bone marrow biopsy confirmed PMF. The patient's recovery was uneventful, with hematological parameters stabilized upon discharge. The case emphasizes the importance of considering myeloproliferative neoplasms (MPNs) in the differential diagnosis of unexplained thrombotic events. JAK2 mutations are linked to thrombotic complications and aneurysm formation, highlighting the need for vigilant monitoring. It is also important that MPNs may not initially be evident in a complete blood count, while coexisting conditions, such as β-thalassemia trait in this patient's case, can alter the blood count findings.
Collapse
Affiliation(s)
- Konstantinos Manganas
- First Department of Propaedeutic and Internal Medicine, Laiko General Hospital, Athens, GRC
| | | | - Klairi Papachristou
- First Department of Propaedeutic and Internal Medicine, Laiko General Hospital, Athens, GRC
| | - Maria Angelara
- First Department of Propaedeutic and Internal Medicine, Laiko General Hospital, Athens, GRC
| | - George Karamanakos
- First Department of Propaedeutic and Internal Medicine, Laiko General Hospital, Athens, GRC
| |
Collapse
|
11
|
Agarwal S, Qamar U, Khan MS, Al-Juhaishi T, Naqash AR, Guha A, Yang EH, Barac A, Ul Abideen Asad Z. Trends and disparities in cardiovascular disease-related mortality among adults with myeloproliferative neoplasms in USA. EUROPEAN HEART JOURNAL OPEN 2025; 5:oeae101. [PMID: 39781540 PMCID: PMC11707680 DOI: 10.1093/ehjopen/oeae101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/02/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025]
Abstract
Aims We aimed to perform a retrospective cohort study using the Centers for Disease Control and Prevention's (CDC's) Wide-Ranging Online Data for Epidemiologic Research (WONDER) database to analyse the trends in cardiovascular disease (CVD)-related mortality in patients with myeloproliferative neoplasms (MPNs) from 1999 to 2020. Methods and results We analysed the death certificate data from the CDC WONDER database from 1999 to 2020 for CVD with co-morbid myeloproliferative disorders in the US population. Age-adjusted mortality rates (AAMRs) and 95% confidence intervals (CIs) were computed per 1 million population by standardizing crude mortality rates to the 2000 US census population. To assess annual national mortality trends, we employed the Joinpoint regression model, calculating the annual per cent change in AAMR and corresponding 95% CIs. A total of 15 269 deaths related to CVD occurred in patients with co-morbid MPNs from 1999 to 2020. Overall, there was a decreasing trend in CVD-related AAMRs throughout these years. Males contributed to 51% of total deaths, and their AAMR was persistently higher than women throughout the study. Non-Hispanic (NH) Whites had the highest overall AAMR, followed by NH Blacks, NH American Indians or Alaska Natives, Hispanics or Latinos, and NH Asian or Pacific Islanders. Conclusion Our findings indicate a significant decline with notable gender, racial/ethnic, and regional differences in CVD-related mortality among patients with MPN over the past two decades. We emphasize the importance of a collaborative approach between oncologists and cardiologists in managing these patients, highlighting the potential benefits of integrating cardio-oncology services to enhance patient outcomes.
Collapse
Affiliation(s)
- Siddharth Agarwal
- Department of Cardiovascular Disease, Mayo Clinic, Rochester, MN 55905, USA
| | - Usama Qamar
- Department of Medicine, King Edward Medical University, Lahore 54000, Pakistan
| | - Muhammad Shahzeb Khan
- Department of Cardiovascular Disease, Duke University School of Medicine, Durham, NC 27710, USA
| | - Taha Al-Juhaishi
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 63117, USA
| | - Abdul Rafeh Naqash
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 63117, USA
| | - Avirup Guha
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Ana Barac
- Inova Schar Cancer Institute and Inova Heart and Vascular Institute, Fairfax, VA 22042, USA
| | - Zain Ul Abideen Asad
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 63117, USA
| |
Collapse
|
12
|
Blaes A, Nohria A, Armenian S, Bergom C, Thavendiranathan P, Barac A, Sanchez-Petitto G, Desai S, Zullig LL, Morgans AK, Herrmann J. Cardiovascular Considerations After Cancer Therapy: Gaps in Evidence and JACC: CardioOncology Expert Panel Recommendations. JACC CardioOncol 2025; 7:1-19. [PMID: 39896126 PMCID: PMC11782100 DOI: 10.1016/j.jaccao.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 02/04/2025] Open
Abstract
Cancer survivors, particularly those treated with anthracyclines and chest radiation, face an elevated risk of cancer therapy-related cardiovascular toxicity. These complications affect not only physical health, but also life expectancy. Risk factors for cancer therapy-related cardiovascular toxicity include age at which cancer treatment was received, the use of (potentially) cardiotoxic cancer therapies, and the presence of concomitant cardiovascular risk factors. Current guidelines provide recommendations for cardiovascular surveillance after cancer therapy, including type and frequency. All cancer survivors are advised to undergo annual clinical screenings and optimization of cardiovascular risk factors. Those at higher risk should undergo additional cardiovascular testing. This document aims to summarize the available evidence, present practical recommendations, and outline existent gaps in the current literature regarding cardiovascular care after cancer therapies.
Collapse
Affiliation(s)
- Anne Blaes
- Division of Hematology/Oncology/Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anju Nohria
- Cardio-Oncology Program, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Saro Armenian
- Department of Pediatrics, City of Hope, Duarte, California, USA
| | - Carmen Bergom
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Paaladinesh Thavendiranathan
- Ted Rogers Program in Cardiotoxicity Prevention, Division of Cardiology, Peter Munk Cardiac Center, University of Toronto, Toronto, Ontario, Canada
| | - Ana Barac
- Inova Schar Heart and Vascular and Inova Schar Cancer Institute, Falls Church, Virginia, USA
| | | | - Sanjal Desai
- Division of Hematology/Oncology/Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Leah L. Zullig
- Department of Population Health Sciences, Duke University, Durham, North Carolina, USA
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham VA Health Care System, Durham, North Carolina, USA
| | | | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
13
|
Amin T, Rasool MHU, Ozkan BI, Swaminathan G, Rauf F, Patrizi S, Sethi A, Frishman WH, Aronow WS, Ahmed MS. Leukocytosis as a Risk Factor for Coronary Artery Disease: Pathophysiology and Epidemiology. Cardiol Rev 2024. [DOI: 10.1097/crd.0000000000000824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Coronary artery disease (CAD) is a significant health concern characterized by reduced blood flow to the heart muscle, primarily due to the buildup of atherosclerotic plaques in the coronary arteries. This process begins with endothelial injury, leading to a cascade of biological responses contributing to plaque formation. Endothelial injury attracts the migration of monocytes which differentiate into macrophages upon uptake of oxidized low-density lipoproteins, changing into lipid-laden macrophage or “foam cells.” The process of plaque formation is influenced by many factors which have been studied extensively in literature such as smoking, hypertension, and diabetes mellitus. Chronic inflammatory illnesses are often associated with a high prevalence of coronary artery syndromes, prompting the evaluation of markers of inflammation such as white blood cell count and inflammatory markers as independent risk factors for CAD. White blood cells play a remarkable role in the pathophysiology of disease formation and progression. The article below aims to discuss the pathophysiology and epidemiology of leukocytosis as a risk factor for CAD.
Collapse
Affiliation(s)
- Toka Amin
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | | | - Bike Ilyada Ozkan
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Gowri Swaminathan
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Faateh Rauf
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Santino Patrizi
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Arshia Sethi
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | | | - Wilbert S. Aronow
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Mahmoud Samy Ahmed
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
14
|
Drofenik A, Blinc A, Bozic Mijovski M, Pajic T, Vrtovec M, Sever M. Relation of JAK2 V617F allele burden and coronary calcium score in patients with essential thrombocythemia. Radiol Oncol 2024; 58:565-572. [PMID: 39361963 PMCID: PMC11604290 DOI: 10.2478/raon-2024-0036] [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: 03/24/2024] [Accepted: 06/01/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND JAK2 V617F (JAK2) mutation is associated with clonal hemopoiesis in myeloproliferative neoplasms as well as with faster progression of cardiovascular diseases. Little is known about the relationship between allele burden and the degree of atherosclerotic alteration of coronary vasculature. We previously reported that carotid artery stiffness progressed faster in patients with JAK2 positive essential thromocythemia (ET) patients. After a four-year follow-up we investigated whether mutation burden of a JAK2 allele correlates with a higher coronary calcium score. PATIENTS AND METHODS Thirty-six patients with JAK2 positive ET and 38 healthy matched control subjects were examined twice within four years. At each visit clinical baseline characteristics and laboratory testing were performed, JAK2 mutation burden was determined, and coronary calcium was measured. RESULTS JAK2 allele burden decreased in 19 patients, did not change in 5 patients, and increased in 4 patients. The coronary calcium Agatston score increased slightly in both groups. Overall, there was no correlation between JAK2 allele burden and calcium burden of coronary arteries. However, in patients with the JAK2 mutation burden increase, the coronary calcium score increased as well. CONCLUSIONS The average JAK2 allele burden decreased in our patients with high-risk ET during the four-year period. However, in the small subgroup whose JAK2 mutation burden increased the Agatston coronary calcium score increased as well. This finding, which should be interpreted with caution and validated in a larger group, is in line with emerging evidence that JAK2 mutation accelerates atherosclerosis and can be regarded as a non-classical risk factor for cardiovascular disease.
Collapse
Affiliation(s)
- Ajda Drofenik
- Department of Cardiology, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Ales Blinc
- Department of Vascular Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mojca Bozic Mijovski
- Department of Vascular Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Tadej Pajic
- Clinical Institute for Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Matjaz Vrtovec
- Department of Vascular Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Department of Dermatovenerology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Matjaz Sever
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Haematology, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
15
|
Momi S, Gresele P. The Role of Platelets in Atherosclerosis: A Historical Review. Semin Thromb Hemost 2024. [PMID: 39561814 DOI: 10.1055/s-0044-1795097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Atherosclerosis is a chronic, multifactorial inflammatory disorder of large and medium-size arteries, which is the leading cause of cardiovascular mortality and morbidity worldwide. Although platelets in cardiovascular disease have mainly been studied for their crucial role in the thrombotic event triggered by atherosclerotic plaque rupture, over the last two decades it has become clear that platelets participate also in the development of atherosclerosis, owing to their ability to interact with the damaged arterial wall and with leukocytes. Platelets participate in all phases of atherogenesis, from the initial functional damage to endothelial cells to plaque unstabilization. Platelets deposit at atherosclerosis predilection sites before the appearance of manifest lesions to the endothelium and contribute to induce endothelial dysfunction, thus supporting leukocyte adhesion to the vessel wall. In particular, platelets release matrix metalloproteinases, which interact with protease-activated receptor 1 on endothelial cells triggering adhesion molecule expression. Moreover, P-selectin and glycoprotein Ibα expressed on the surface of vessel wall-adhering platelets bind PSGL-1 and β2 integrins on leukocytes, favoring their arrest and transendothelial migration. Platelet-leukocyte interactions promote the formation of radical oxygen species which are strongly involved in the lipid peroxidation associated with atherosclerosis. Platelets themselves actively migrate through the endothelium toward the plaque core where they release chemokines that modify the microenvironment by modulating the function of other inflammatory cells, such as macrophages. While current antiplatelet agents seem unable to prevent the contribution of platelets to atherogenesis, the inhibition of platelet secretion, of the release of MMPs, and of some specific pathways of platelet adhesion to the vessel wall may represent promising future strategies for the prevention of atheroprogression.
Collapse
Affiliation(s)
- Stefania Momi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Gresele
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
16
|
Di Vito L, Di Giusto F, Mazzotta S, Scalone G, Bruscoli F, Silenzi S, Selimi A, Angelini M, Galieni P, Grossi P. Management of vulnerable patient phenotypes and acute coronary syndrome mechanisms. Int J Cardiol 2024; 415:132365. [PMID: 39029561 DOI: 10.1016/j.ijcard.2024.132365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/07/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Atherosclerosis is a chronic vascular disease. Its prevalence increases with aging. However, atherosclerosis may also affect young subjects without significant exposure to the classical risk factors. Recent evidence indicates clonal hematopoiesis of indeterminate potential (CHIP) as a novel cardiovascular risk factor that should be suspected in young patients. CHIP represents a link between impaired bone marrow and atherosclerosis. Atherosclerosis may present with an acute symptomatic manifestation or subclinical events that favor plaque growth. The outcome of a plaque relies on a balance of innate and environmental factors. These factors can influence the processes that initiate and propagate acute plaque destabilization leading to intraluminal thrombus formation or subclinical vessel healing. Thirty years ago, the first autopsy study revealed that coronary plaques can undergo rupture even in subjects without a known cardiovascular history. Nowadays, cardiac magnetic resonance studies demonstrate that this phenomenon is not rare. Myocardial infarction is mainly due to plaque rupture and plaque erosion that have different pathophysiological mechanisms. Plaque erosion carries a better prognosis as compared to plaque rupture. Thus, a tailored conservative treatment has been proposed and some studies demonstrated it to be safe. On the contrary, plaque rupture is typically associated with inflammation and anti-inflammatory treatments have been proposed in response to persistently elevate biomarkers of systemic inflammation. In conclusion, atherosclerosis may present in different forms or phenotypes. Vulnerable patient phenotypes, identified by using intravascular imaging techniques, biomarkers, or even genetic analyses, are characterized by distinctive pathophysiological mechanisms. These different phenotypes merit tailored management.
Collapse
Affiliation(s)
- Luca Di Vito
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy.
| | | | - Serena Mazzotta
- Department of Haematology and Stem Cell Transplantation Unit C. e G, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Giancarla Scalone
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy
| | - Filippo Bruscoli
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy
| | - Simona Silenzi
- Cardiology Unit, C. and G, Mazzoni Hospital, AST Ascoli Piceno, Italy
| | - Adelina Selimi
- University Hospital "Umberto I-Lancisi-Salesi", Ancona, Italy
| | - Mario Angelini
- Department of Haematology and Stem Cell Transplantation Unit C. e G, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Piero Galieni
- Department of Haematology and Stem Cell Transplantation Unit C. e G, Mazzoni Hospital, Ascoli Piceno, Italy
| | | |
Collapse
|
17
|
Mukherjee P, Ansell SM, Mondello P. Unraveling the role of cancer-associated fibroblasts in B cell lymphoma. Front Immunol 2024; 15:1451791. [PMID: 39555055 PMCID: PMC11563820 DOI: 10.3389/fimmu.2024.1451791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/03/2024] [Indexed: 11/19/2024] Open
Abstract
Recent breakthroughs in research have sparked a paradigm shift in our understanding of cancer biology, uncovering the critical role of the crosstalk between tumor cells and the immune cells of the tumor microenvironment (TME) in malignant transformation. Fibroblasts have long been viewed as ancillary participants in cancer progression, often eclipsed by the prominence given to malignant cells. Novel investigations, however, have increasingly acknowledged the essential part played by the fibroblasts and their phenotypic doppelganger cancer-associated fibroblasts (CAFs) in fostering immunosuppression and promoting tumor progression. Here we review the cell-of-origin from which CAFs derive and their altered programs compared to their normal counterpart. We will also discuss the complex interplay between CAFs and the surrounding immune cells of the TME in the context of solid tumors and B cell lymphomas, with a focus on the "reprogrammable" role of CAFs in immunosuppression, immuno-activation and immuno-avoidance, and their implications on drug resistance. Finally, we will examine the existing and plausible therapeutic approaches targeting CAFs as a strategy to enhance treatment response.
Collapse
|
18
|
Tang M, Chen Y, Zhou Y, Zhuang X, Fu Y, Chen J, Wei R, Chen Y. Long-term risks of cardiovascular-specific mortality among myeloproliferative neoplasms patients. Ther Adv Hematol 2024; 15:20406207241290886. [PMID: 39478808 PMCID: PMC11523155 DOI: 10.1177/20406207241290886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024] Open
Abstract
Background The myeloproliferative neoplasm (MPN) is a heterogeneous group of clonal hyperplasia hematopoietic stem cell disorders, predominantly affecting middle-aged and elderly individuals, with a slow disease progression. With advancements in disease-related research, the survival rates of MPN patients have significantly improved. This research primarily focuses on cardiovascular disease mortality (CVM) and prognostic factors in MPN patients, aiming to provide clinicians with more comprehensive references. Methods A total of 24,277 patients were included in the Surveillance, Epidemiology, and End Results (SEER) database. Cumulative mortality was assessed using a competing risk model, univariate and multivariate regression analysis of cardiovascular disease (CVD) mortality risk factors, and a comparison of standardized mortality ratio (SMR) and general population CVM. Results Among the 24,277 patients included in this study, a total of 8841 deaths occurred during the follow-up period, with 2429 attributed to CVD. Notably, the risk of CVM was found to be significantly higher in patients with MPNs compared to the general population. Furthermore, this risk increased over time. CVD emerged as the predominant cause of death among individuals aged over 80 years and younger patients exhibited a significantly elevated SMR. Additionally, age, race, marital status, and insurance status were identified as independent prognostic factors for CVM. Conclusion The incidence of cardiovascular events in patients with MPNs is significantly higher compared to the general population. Early screening and assessment of cardiac health should be implemented in MPN patients to prevent the occurrence of cardiovascular events and enhance their prognosis.
Collapse
Affiliation(s)
- Meiling Tang
- Department of Hematology, The Eighth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Meiling Tang is currently affiliated to Department of Pulmonary and Critical Care Medicine, the Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou, China
- Ying Chen is currently affiliated to Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ying Chen
- Department of Hematology, The Eighth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Meiling Tang is currently affiliated to Department of Pulmonary and Critical Care Medicine, the Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou, China
- Ying Chen is currently affiliated to Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yanying Zhou
- Department of Hematology, The Eighth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Meiling Tang is currently affiliated to Department of Pulmonary and Critical Care Medicine, the Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou, China
- Ying Chen is currently affiliated to Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xinran Zhuang
- Department of Hematology, The Eighth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Meiling Tang is currently affiliated to Department of Pulmonary and Critical Care Medicine, the Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou, China
- Ying Chen is currently affiliated to Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuxin Fu
- Department of Hematology, The Eighth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Meiling Tang is currently affiliated to Department of Pulmonary and Critical Care Medicine, the Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou, China
- Ying Chen is currently affiliated to Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jinzheng Chen
- Department of Hematology, The Eighth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Meiling Tang is currently affiliated to Department of Pulmonary and Critical Care Medicine, the Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou, China
- Ying Chen is currently affiliated to Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Rongfang Wei
- Department of Hematology, The Eighth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Meiling Tang is currently affiliated to Department of Pulmonary and Critical Care Medicine, the Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou, China
- Ying Chen is currently affiliated to Medical Center of Hematology, State Key Laboratory of Trauma, Burn and Combined Injury, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yan Chen
- Department of Hematology, The Eighth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, 3025 Shennan Middle Road, Shenzhen 518000, China
| |
Collapse
|
19
|
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
|
20
|
Tiu A, McKinnell Z, Liu S, Gill P, Antonio M, Shancer Z, Srinivasa N, Diao G, Subrahmanyam R, Kessler CM, Jain M. Risk of myeloproliferative neoplasms among U.S. Veterans from Korean, Vietnam, and Persian Gulf War eras. Am J Hematol 2024; 99:1969-1978. [PMID: 39023278 DOI: 10.1002/ajh.27438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
The Promise to Address Comprehensive Toxics (PACT) Act expanded U.S. Veterans' health care and benefits for conditions linked to service-connected exposures (e.g., Burn Pits, Agent Orange). However, myeloproliferative neoplasms (MPN) are not recognized as presumptive conditions for Veterans exposed to these toxic substances. This study evaluated the development of MPN among U.S. Veterans from the Korean, Vietnam, and Persian Gulf War eras. This retrospective cohort study included 65 425 Korean War era Veterans; 211 927 Vietnam War era Veterans; and 214 007 Persian Gulf War era Veterans from January 1, 2006, to January 26, 2023. Veterans with MPN, thrombosis, bleeding, and cardiovascular risk factors were identified through ICD-9 and -10 codes. Veterans from the Persian Gulf War era had the highest risk of developing MPN compared with Veterans from the Korean and Vietnam War eras, hazard ratio (HR) 4.92, 95% confidence interval (CI) 4.20-5.75 and HR 2.49, 95% CI 2.20-2.82, both p < .0001, respectively. Vietnam War era Veterans also had a higher risk of MPN development compared with Korean War era Veterans, HR 1.97, 95% CI 1.77-2.21, p < .0001. Persian Gulf War era Veterans were diagnosed with MPN at an earlier age, had higher risks of thrombosis and bleeding, and had lower survival rates compared with Korean War and Vietnam War era Veterans. This study reinforces evidence that environmental and occupational hazards increase the risk of clonal myeloid disorders and related complications, impacting overall survival with MPN. Limitations include the inability to confirm clonality and fully verify deployment and exposure status.
Collapse
Affiliation(s)
- Andrew Tiu
- Division of Hematology-Oncology, Lombardi Comprehensive Cancer Center, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Zoe McKinnell
- Division of Hematology-Oncology, The George Washington University, Washington, DC, USA
| | - Shanshan Liu
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Puneet Gill
- Institute for Clinical Research, Washington, DC, USA
| | | | - Zoe Shancer
- The George Washington University School of Medicine, Washington, DC, USA
| | - Nandan Srinivasa
- The George Washington University School of Medicine, Washington, DC, USA
| | - Guoqing Diao
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | | | - Craig M Kessler
- Division of Hematology-Oncology, Lombardi Comprehensive Cancer Center, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Maneesh Jain
- The George Washington University School of Medicine, Washington, DC, USA
- Washington DC VA Medical Center, Washington, DC, USA
| |
Collapse
|
21
|
Gajagowni S, Hopkins S, Qadeer Y, Virani SS, Verdonschot JAJ, Coombs CC, Amos CI, Nead KT, Jaiswal S, Krittanawong C. Clonal hematopoiesis of indeterminate potential and cardiovascular disease: Pathogenesis, clinical presentation, and future directions. Prog Cardiovasc Dis 2024; 86:79-85. [PMID: 39278303 DOI: 10.1016/j.pcad.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is a well-studied phenomenon in hematologic malignancies. With advancements in gene sampling and analysis and the use of large cohort studies, CHIP has recently been linked to cardiovascular disease (CVD). The relationship between CHIP and CVD appears to be bidirectional, with traditional risk factors for cardiovascular disease increasing the mutation burden in CHIP, and CHIP itself effecting the incidence or prognosis of a variety of CVD. The purpose of this review is to understand the epidemiology, risk factors, and pathogenesis of CHIP in the context of various CVD conditions.
Collapse
Affiliation(s)
- Saivaroon Gajagowni
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Steven Hopkins
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Yusuf Qadeer
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Salim S Virani
- Office of the Vice Provost (Research), The Aga Khan University, Karachi 74800, Pakistan; Section of Cardiology and Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Job A J Verdonschot
- Department of Cardiology, Maastricht University Medical Centre, Center for Heart Failure Research, Cardiovascular Research Institute Maastricht (CARIM), University Hospital Maastricht, P. Debyelaan 25, 6229, HX, Maastricht, the Netherlands; Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW), Maastricht University Medical Center, 6202, AZ, Maastricht, the Netherlands
| | - Catherine C Coombs
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, California, United States of America
| | - Christopher I Amos
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Kevin T Nead
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, United States of America; Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Siddhartha Jaiswal
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Chayakrit Krittanawong
- Cardiology Division, NYU Langone Health and NYU School of Medicine, New York, NY, United States of America.
| |
Collapse
|
22
|
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
|
23
|
Magyar E, Újfalusi A, Czenke M, Méhes G. [Somatic JAK2 gene mutation is an evolving risk factor in cardiovascular diseases]. Orv Hetil 2024; 165:883-890. [PMID: 38852144 DOI: 10.1556/650.2024.33055] [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: 03/14/2024] [Accepted: 04/24/2024] [Indexed: 06/11/2024]
Abstract
A JAK-kináz működése egyes, sejtfelszíni receptorokhoz kötött jelátviteli
útvonalak központi eleme (JAK/STAT útvonal), a fiziológiás sejtaktiváció
közvetítője. Közvetítő hatása a myeloid eredetű sejtekben, elsősorban
macrophagokban, neutrophil granulocytákban, illetve a thrombocytákban is
szükséges a nem specifikus gyulladásos reakció elindításához. A jelátviteli
útvonal kóros aktivációja a krónikus myeloproliferativ neoplasiákban az egyik
leggyakoribb genetikai eltérés és a fokozott myelo- és thrombopoesis egyik jól
ismert ’driver’ mechanizmusa. A myeloproliferativ neoplasiák diagnosztikájához a
szerzett patogén JAK2-mutációk (elsősorban a
V617F variáns) kimutatása is hozzátartozik. Ismertté vált
ugyanakkor, hogy a génhiba – más, ritkább eltérések mellett – akár hosszú ideig
fennállhat jelentős hematológiai eltérések nélkül. Ezt az állapotot klonális
haemopoesisnek nevezik. A variáns JAK2 újabb megfigyelések
szerint a myeloid eredetű sejtek funkciójára ebben a helyzetben is komoly
aktiváló hatást fejthet ki, és meggyőző összefüggéseket mutattak ki a
gyulladásos háttérrel rendelkező cardiovascularis szövődményekkel kapcsolatosan.
A klonális JAK2-eltérés, valamint az ischaemiás szívbetegség,
illetve a hasi aortaaneurysma kialakulása között is jelentős összefüggések
mérhetők. Mára egyértelmű, hogy az atheromaképződés és a következményes
coronariasclerosis kialakulása egy komplex thromboinflammatoricus folyamat
következménye, és jelentős mértékben függ többek között a JAK2
mediálta szöveti macrophag–granulocyta interakcióktól. Közleményünkben a
fokozott kockázat hátterében húzódó mechanizmusokat és a rendelkezésre álló
legújabb ismereteket elemezzük. Orv Hetil. 2024; 165(23): 883–890.
Collapse
Affiliation(s)
- Eszter Magyar
- 1 Debreceni Egyetem, Általános Orvostudományi Kar, Patológiai Intézet Debrecen, Nagyerdei krt. 98., 4032 Magyarország
| | - Anikó Újfalusi
- 2 Debreceni Egyetem, Általános Orvostudományi Kar, Laboratóriumi Medicina Intézet Budapest Magyarország
| | - Marianna Czenke
- 1 Debreceni Egyetem, Általános Orvostudományi Kar, Patológiai Intézet Debrecen, Nagyerdei krt. 98., 4032 Magyarország
| | - Gábor Méhes
- 1 Debreceni Egyetem, Általános Orvostudományi Kar, Patológiai Intézet Debrecen, Nagyerdei krt. 98., 4032 Magyarország
| |
Collapse
|
24
|
Wen S, Zhang W, Fei Y, Guan K, Zhao H, Song P, Ye X, Pan Y. Risk factors for ischemic stroke in patients with Philadelphia chromosome (Ph)-negative myeloproliferative neoplasms. J Clin Neurosci 2024; 125:159-166. [PMID: 38815302 DOI: 10.1016/j.jocn.2024.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Philadelphia chromosome-negative myeloproliferative neoplasms (Ph-negative MPNs) are linked with various complications, notably ischemic stroke. The study aims to identify risk factors for ischemic stroke in Ph-negative MPNs patients. METHODS Patients were categorized into two groups based on whether they had experienced ischemic stroke. Subsequently, an analysis of demographics, biochemical makers, and genetic mutations (JAK2V617F and CALR mutations), was conducted to identify potential associations with an elevated risk of ischemic stroke in individuals with Ph-negative MPNs. RESULTS A total of 185 patients diagnosed with Ph-negative MPNs participated in the study, including 82 with essential thrombocythemia (ET), 78 with polycythemia vera (PV), and 25 with primary myelofibrosis (PMF). Among these, 57 patients (30.8 %) had a history of ischemic stroke. Independent risk factors associated with ischemic stroke in Ph-negative MPNs patients included hypertension (OR = 5.076) and smoking (OR = 5.426). Among ET patients, smoking (OR = 4.114) and an elevated percentage of neutrophils (OR = 1.080) were both positively correlated with ischemic stroke incidence. For PV patients, hypertension (OR = 4.647), smoking (OR = 6.065), and an increased percentage of lymphocytes (OR = 1.039) were independently associated with ischemic stroke. Regardless of the presence of the JAK2V617F mutation, hypertension was the sole positively and independently associated risk factor for ischemic stroke. The odds ratios for patients with the JAK2V617F mutation was 3.103, while for those without the mutation, it was 11.25. CONCLUSIONS Hypertension was a more substantial factor associated with an increased incidence of ischemic stroke in Ph-negative MPNs patients.
Collapse
Affiliation(s)
- Shirong Wen
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wenxiao Zhang
- Department of Neurology, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, China
| | - Yiping Fei
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ke Guan
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Hui Zhao
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Peng Song
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiangmei Ye
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yujun Pan
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
25
|
Lucijanic M, Krecak I, Soric E, Sabljic A, Galusic D, Holik H, Perisa V, Moric Peric M, Zekanovic I, Budimir J, Kusec R. Evaluation of Absolute Neutrophil, Lymphocyte and Platelet Count and Their Ratios as Predictors of Thrombotic Risk in Patients with Prefibrotic and Overt Myelofibrosis. Life (Basel) 2024; 14:523. [PMID: 38672793 PMCID: PMC11051164 DOI: 10.3390/life14040523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
AIM To investigate the prognostic contribution of absolute neutrophil (ANC), lymphocyte (ALC), platelet count and their ratios, neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR), to thrombotic risk in patients with prefibrotic and overt fibrotic myelofibrosis (MF). METHODS We retrospectively analyzed a cohort of 256 patients with prefibrotic (85 patients) and overt fibrotic MF (171 patients) treated in six Croatian hematological centers. RESULTS Prefibrotic compared to overt fibrotic MF patients presented with significantly higher ALC, platelet count and PLR, and experienced longer time to thrombosis (TTT). Among prefibrotic patients, ANC > 8.33 × 109/L (HR 13.08, p = 0.036), ALC > 2.58 × 109/L (HR 20.63, p = 0.049) and platelet count > 752 × 109/L (HR 10.5, p = 0.043) remained independently associated with shorter TTT. Among overt fibrotic patients, ANC > 8.8 × 109/L (HR 4.49, p = 0.004), ALC ≤ 1.43 × 109/L (HR 4.15, p = 0.003), platelet count ≤ 385 × 109/L (HR 4.68, p = 0.004) and chronic kidney disease (HR 9.07, p < 0.001) remained independently associated with shorter TTT. CONCLUSIONS Prognostic properties of ANC, ALC and platelet count are mutually independent and exceed those of NLR and PLR regarding thrombotic risk stratification. ALC and platelet count associate in opposite directions with thrombotic risk in prefibrotic and overt fibrotic MF patients.
Collapse
Affiliation(s)
- Marko Lucijanic
- Hematology Department, University Hospital Dubrava, Av. Gojka Suska 6, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, Ul. Salata 3, 10000 Zagreb, Croatia
| | - Ivan Krecak
- Department of Internal Medicine, General Hospital Sibenik, Ul. Stjepana Radica 83, 22000 Sibenik, Croatia
- School of Medicine, University of Rijeka, Ul. Brace Branchetta 20/1, 51000 Rijeka, Croatia
- Sibenik University of Applied Science, Trg Andrije Hebranga 11, 22000 Sibenik, Croatia
| | - Ena Soric
- Hematology Department, University Hospital Dubrava, Av. Gojka Suska 6, 10000 Zagreb, Croatia
| | - Anica Sabljic
- Hematology Department, University Hospital Dubrava, Av. Gojka Suska 6, 10000 Zagreb, Croatia
| | - Davor Galusic
- Department of Hematology, University Hospital of Split, Soltanska ul. 1, 21000 Split, Croatia
- School of Medicine, University of Split, Soltanska ul. 2, 21000 Split, Croatia
| | - Hrvoje Holik
- Department of Internal Medicine, “Dr. Josip Bencevic” General Hospital, Ul. Andrije Stampara, 35000 Slavonski Brod, Croatia
- Faculty of Medicine, University of Osijek, Ul. Josipa Huttlera 4, 31000 Osijek, Croatia
| | - Vlatka Perisa
- Faculty of Medicine, University of Osijek, Ul. Josipa Huttlera 4, 31000 Osijek, Croatia
- Department of Hematology, Osijek University Hospital, Ul. Josipa Huttlera 4, 31000 Osijek, Croatia
| | - Martina Moric Peric
- Department of Internal Medicine, General Hospital Zadar, Ul. Boze Pericica 5, 23000 Zadar, Croatia
| | - Ivan Zekanovic
- Department of Internal Medicine, General Hospital Zadar, Ul. Boze Pericica 5, 23000 Zadar, Croatia
| | - Josipa Budimir
- Department of Internal Medicine, General Hospital Sibenik, Ul. Stjepana Radica 83, 22000 Sibenik, Croatia
| | - Rajko Kusec
- Hematology Department, University Hospital Dubrava, Av. Gojka Suska 6, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, Ul. Salata 3, 10000 Zagreb, Croatia
| |
Collapse
|
26
|
Leiva O, How J, Grevet J, Brunner A, Hobbs G. In-Hospital and readmission outcomes of patients with myeloproliferative neoplasms and atrial fibrillation: insights from the National Readmissions Database. J Thromb Thrombolysis 2024; 57:186-193. [PMID: 37839025 DOI: 10.1007/s11239-023-02900-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
INTRODUCTION Patients with myeloproliferative neoplasms (MPNs) and atrial fibrillation (AF) are at increased risk of thrombosis and bleeding. However, the risk of thrombosis and bleeding in patients with AF and MPN compared with the general population with AF is unclear. Additionally, traditional risk scores (CHA2DS2-VASC and HAS-BLED) for risk/benefit estimation of thromboprophylaxis in AF do not account for MPN status. Therefore, we aimed to investigate bleeding and thrombosis risk in patients with MPN hospitalized for AF. METHODS We utilized the National Readmission Database (NRD) to identify patients with AF with and without MPN. Primary bleeding and thrombosis outcomes were in-hospital or 30-day readmission for bleeding or thrombosis, respectively. We propensity score (PS) matched patients with and without MPN. Risk of primary outcomes in MPN was assessed in PS matched cohort using logistic regression. Receiver operating characteristic (ROC) curve used to evaluate predictive ability of CHA2DS2-VASC and HAS-BLED of primary thrombosis and bleeding outcomes, respectively. RESULTS 24,185 patients without MPN were matched with 1,617 patients with MPN and variables were balanced between groups. Patients with MPN were at increased risk of meeting the thrombosis (OR 1.98, 95% CI 1.23-3.21) but not bleeding (OR 0.87, 95% CI 0.63-1.19) primary outcomes. In MPN, CHA2DS2-VASC predicted thrombosis (C-statistic 0.66, 95% CI 0.54-0.78) but HAS-BLED was a poor predictor of bleeding (C-statistic 0.55, 95% CI 0.46-0.64). CONCLUSION In patients with AF, MPN was associated with increased risk of bleeding and thrombosis. HAS-BLED scores did not accurately predict bleeding in MPN. Further investigation is needed to refine risk scores in MPN.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Joan How
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Jeremy Grevet
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew Brunner
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, USA
| | - Gabriela Hobbs
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, USA.
| |
Collapse
|
27
|
Liu C, Zhou YP, Lian TY, Li RN, Ma JS, Yang YJ, Zhang SJ, Li XM, Qiu LH, Qiu BC, Ren LY, Wang J, Han ZY, Li JH, Wang L, Xu XQ, Sun K, Chen LF, Cheng CY, Zhang ZJ, Jing ZC. Clonal Hematopoiesis of Indeterminate Potential in Chronic Thromboembolic Pulmonary Hypertension: A Multicenter Study. Hypertension 2024; 81:372-382. [PMID: 38116660 DOI: 10.1161/hypertensionaha.123.22274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND The pathogenesis of chronic thromboembolic pulmonary hypertension (CTEPH) is multifactorial and growing evidence has indicated that hematological disorders are involved. Clonal hematopoiesis of indeterminate potential (CHIP) has recently been associated with an increased risk of both hematological malignancies and cardiovascular diseases. However, the prevalence and clinical relevance of CHIP in patients with CTEPH remains unclear. METHODS Using stepwise calling on next-generation sequencing data from 499 patients with CTEPH referred to 3 centers between October 2006 and December 2021, CHIP mutations were identified. We associated CHIP with all-cause mortality in patients with CTEPH. To provide insights into potential mechanisms, the associations between CHIP and inflammatory markers were also determined. RESULTS In total, 47 (9.4%) patients with CTEPH carried at least 1 CHIP mutation at a variant allele frequency of ≥2%. The most common mutations were in DNMT3A, TET2, RUNX1, and ASXL1. During follow-up (mean, 55 months), deaths occurred in 22 (46.8%) and 104 (23.0%) patients in the CHIP and non-CHIP groups, respectively (P<0.001, log-rank test). The association of CHIP with mortality remained robust in the fully adjusted model (hazard ratio, 2.190 [95% CI, 1.257-3.816]; P=0.006). Moreover, patients with CHIP mutations showed higher circulating interleukin-1β and interleukin-6 and lower interleukin-4 and IgG galactosylation levels. CONCLUSIONS This is the first study to show that CHIP mutations occurred in 9.4% of patients with CTEPH are associated with a severe inflammatory state and confer a poorer prognosis in long-term follow-up.
Collapse
Affiliation(s)
- Chao Liu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-Ping Zhou
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian-Yu Lian
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China (T.-Y.L., S.-J.Z., C.-Y.C., Z.-C.J)
| | - Ruo-Nan Li
- School of Pharmacy, Henan University, Zhengzhou, China (R.-N.L., J.-S.M.)
| | - Jing-Si Ma
- School of Pharmacy, Henan University, Zhengzhou, China (R.-N.L., J.-S.M.)
| | - Yin-Jian Yang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (Y.-J.Y., K.S., Z.-J.Z.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Si-Jin Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China (T.-Y.L., S.-J.Z., C.-Y.C., Z.-C.J)
| | - Xian-Mei Li
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lu-Hong Qiu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bao-Chen Qiu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Yan Ren
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Wang
- Department of Medical Laboratory, Weifang Medical University, China (J.W.)
| | - Zhi-Yan Han
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital (Z.-Y.H., J.-H.L.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Hui Li
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital (Z.-Y.H., J.-H.L.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lan Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University, China (L.W.)
| | - Xi-Qi Xu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Sun
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (Y.-J.Y., K.S., Z.-J.Z.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lian-Feng Chen
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chun-Yan Cheng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China (T.-Y.L., S.-J.Z., C.-Y.C., Z.-C.J)
| | - Ze-Jian Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (Y.-J.Y., K.S., Z.-J.Z.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Cheng Jing
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China (T.-Y.L., S.-J.Z., C.-Y.C., Z.-C.J)
| |
Collapse
|
28
|
Barbui T, Carobbio A, Vannucchi AM, De Stefano V. Are the available data sufficient to suggest cytoreductive agents for patients with CHIP and stroke? Blood Adv 2023; 7:7551-7553. [PMID: 37874911 PMCID: PMC10761349 DOI: 10.1182/bloodadvances.2023012007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 10/26/2023] Open
Affiliation(s)
- Tiziano Barbui
- FROM Research Foundation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | | | - Alessandro M. Vannucchi
- Department of Experimental and Clinical Medicine, Center Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | - Valerio De Stefano
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| |
Collapse
|
29
|
Leiva O, Alvarez-Cardona J, How J, Brunner A, Hobbs G. In-Hospital and readmission outcomes of patients with myeloproliferative neoplasms and heart failure: Insights from the National Readmissions Database. IJC HEART & VASCULATURE 2023; 49:101304. [PMID: 38173785 PMCID: PMC10761308 DOI: 10.1016/j.ijcha.2023.101304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 01/05/2024]
Abstract
Background Myeloproliferative neoplasms (MPNs) are chronic leukemias associated with increased risk of cardiovascular (CV) events. Prior studies suggest patients with MPN are at increased risk of HF. Additionally, pre-clinical murine models harboring the JAK2 mutation, the most common driver mutation in MPNs, have shown accelerated adverse cardiac remodeling in myocardial infarction and pressure overload HF models. However, clinical outcomes, including in-hospital and readmission outcomes, of patients with MPN admitted for HF have not been well characterized. Methods Patients hospitalized for HF with and without MPN were identified using the 2017 and 2018 National Readmission Database. Propensity score matching (PSM) was performed to match 1 MPN with 10 non-MPN controls. Outcomes were in-hospital death, 90-day CV-related, HF-related, and all-cause readmissions. Logistic regression and Cox proportional hazards regression models were used to estimate risk of in-hospital death and 90-day readmission outcomes, respectively. Results After PSM, 4,626 patients with MPN were matched with 46,260 without. Patients with MPN were associated with increased risk of in-hospital death (OR 1.17, 95% CI 1.00 - 1.35), 90-day CV-related (HR 1.10, 95% CI 1.02 - 1.18) and all-cause (HR 1.24, 95% CI 1.17 - 1.31) but not HF-related (HR 1.05, 95% CI 0.97 - 1.14) readmissions. Conclusion Among patients hospitalized for HF, MPN was associated with increased risk of in-hospital death, and 90-day CV-related readmissions (driven primarily by thrombotic readmissions). Further investigation is needed in order to improve outcomes in patients with MPN and HF.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Jose Alvarez-Cardona
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Joan How
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, United States
| | - Andrew Brunner
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Gabriela Hobbs
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
30
|
Manan MR, Kipkorir V, Nawaz I, Waithaka MW, Srichawla BS, Găman AM, Diaconu CC, Găman MA. Acute myocardial infarction in myeloproliferative neoplasms. World J Cardiol 2023; 15:571-581. [PMID: 38058401 PMCID: PMC10696206 DOI: 10.4330/wjc.v15.i11.571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/21/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are a heterogeneous group of hematologic malignancies characterized by an abnormal proliferation of cells of the myeloid lineage. Affected individuals are at increased risk for cardiovascular and thrombotic events. Myocardial infarction (MI) may be one of the earliest clinical manifestations of MPNs or may be a thrombotic complication that develops during the natural course of the disease. In the present review, we examine the epidemiology, pathogenesis, clinical presentation, and management of MI in MPNs based on the available literature. Moreover, we review potential biomarkers that could mediate the MI-MPNs crosstalk, from classical biochemical tests, e.g., lactate dehydrogenase, creatine kinase and troponins, to pro-inflammatory cytokines, oxidative stress markers, and clonal hematopoiesis.
Collapse
Affiliation(s)
| | - Vincent Kipkorir
- Department of Human Anatomy and Physiology, University of Nairobi, Nairobi 00100, Kenya
| | - Iqra Nawaz
- Quaid-e-Azam Medical College, Bahawalpur 63100, Pakistan
| | | | - Bahadar Singh Srichawla
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Amelia Maria Găman
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, Craiova 200143, Romania
- Clinic of Hematology, Filantropia City Hospital, Craiova 200143, Romania
| | - Camelia Cristina Diaconu
- Department of Internal Medicine, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest 050474, Romania
- Internal Medicine Clinic, Clinical Emergency Hospital of Bucharest, Bucharest 105402, Romania
| | - Mihnea-Alexandru Găman
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest 022328, Romania
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, Bucharest 030304, Romania.
| |
Collapse
|
31
|
Skov V, Thomassen M, Kjaer L, Larsen MK, Knudsen TA, Ellervik C, Kruse TA, Hasselbalch HC. Whole blood transcriptional profiling reveals highly deregulated atherosclerosis genes in Philadelphia-chromosome negative myeloproliferative neoplasms. Eur J Haematol 2023; 111:805-814. [PMID: 37640394 DOI: 10.1111/ejh.14081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND The Philadelphia-negative chronic myeloproliferative neoplasms (MPNs) are associated with a huge comorbidity burden, including an increased risk of cardiovascular diseases. Recently, chronic inflammation has been suggested to be the driving force for clonal evolution and disease progression in MPN but also potentially having an impact upon the development of accelerated (premature) atherosclerosis. OBJECTIVES Since chronic inflammation, atherosclerosis, and atherothrombosis are prevalent in MPNs and we have previously shown oxidative stress genes to be markedly upregulated in MPNs, we hypothesized that genes linked to development of atherosclerosis might be highly deregulated as well. METHODS Using whole blood gene expression profiling in patients with essential thrombocythemia (ET; n = 19), polycythemia vera (PV; n = 41), or primary myelofibrosis (PMF; n = 9), we herein for the first time report aberrant expression of several atherosclerosis genes. RESULTS Of 84 atherosclerosis genes, 45, 56, and 46 genes were deregulated in patients with ET, PV, or PMF, respectively. Furthermore, BCL2L1, MMP1, PDGFA, PTGS1, and THBS4 were progressively significantly upregulated and BCL2 progressively significantly downregulated from ET over PV to PMF (all FDR <0.05). CONCLUSIONS We have for the first time shown massive deregulation of atherosclerosis genes in MPNs, likely reflecting the inflammatory state in MPNs in association with in vivo activation of leukocytes, platelets, and endothelial cells being deeply involved in the atherosclerotic process.
Collapse
Affiliation(s)
- Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Lasse Kjaer
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | | | - Trine A Knudsen
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Torben A Kruse
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | | |
Collapse
|
32
|
Kristiansen MH, Kjær L, Skov V, Larsen MK, Ellervik C, Hasselbalch HC, Wienecke T. JAK2V617F mutation is highly prevalent in patients with ischemic stroke: a case-control study. Blood Adv 2023; 7:5825-5834. [PMID: 37522722 PMCID: PMC10561044 DOI: 10.1182/bloodadvances.2023010588] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/01/2023] Open
Abstract
Ischemic stroke has a high recurrence rate despite treatment. This underlines the significance of investigating new possible cerebrovascular risk factors, such as the acquired gene mutation JAK2V617F found in 3.1% of the general population. We aimed to investigate the prevalence of the JAK2V617F mutation in a population with ischemic stroke compared with that in matched controls. We enrolled 538 consecutive Danish patients with ischemic stroke (mean age, 69.5 ± 10.9 years; 39.2% female) within 7 days of symptom onset. Using multiple-adjusted conditional logistic regression analysis, we compared the prevalence of JAK2V617F with that in age- and sex-matched controls free of ischemic cerebrovascular disease (ICVD) from the Danish General Suburban Population Study. DNA was analyzed for JAK2V617F mutation using sensitive droplet digital polymerase chain reaction in patients and controls. Of the 538 patients with ischemic stroke, 61 (11.3%) had JAK2V617F mutation. There were no differences in patient demographics or cerebrovascular comorbidities between the patients with and without mutations. Patients with ischemic stroke were more likely to have the JAK2V617F mutation than matched controls, in whom the JAK2V617F prevalence was 4.4% (odds ratio, 2.37; 95% confidence interval, 1.57-3.58; P < .001). A subanalysis stratified by smoking history revealed that the association was strongest in current smokers (odds ratio, 4.78; 95% confidence interval, 2.22-10.28; P < .001). Patients with ischemic stroke were 2.4 times more likely to have the JAK2V617F mutation than matched controls without ICVD when adjusting for other cerebrovascular risk factors. This finding supports JAK2V617F mutation as a novel cerebrovascular risk factor.
Collapse
Affiliation(s)
- Marie Hvelplund Kristiansen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Morten Kranker Larsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- 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 Data and Data Support, Region Zealand, Sorø, Denmark
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Hans Carl Hasselbalch
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Troels Wienecke
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| |
Collapse
|
33
|
Chandra DJ, Lachowiez CA. Anthracycline induced left ventricular dysfunction in AML: A focus on the molecularly defined future of cardio-oncology. Leuk Res 2023; 133:107366. [PMID: 37531679 DOI: 10.1016/j.leukres.2023.107366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Affiliation(s)
- Daniel J Chandra
- Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Curtis A Lachowiez
- Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
34
|
Leiva O, Garcia BD, Hobbs G. Pulmonary Hypertension in Myeloproliferative Neoplasms: New Insights and Unexplored Horizons. Am J Respir Crit Care Med 2023; 208:518-521. [PMID: 37429036 PMCID: PMC10492239 DOI: 10.1164/rccm.202306-1001ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 07/10/2023] [Indexed: 07/12/2023] Open
Affiliation(s)
- Orly Leiva
- Department of Medicine New York University Grossman School of Medicine New York, New York
| | - Brenda D Garcia
- Department of Medicine Northwell Health-Lenox Hill Hospital and Zucker School of Medicine at Hofstra/Northwell New York, New York
| | - Gabriela Hobbs
- Department of Medicine Massachusetts General Hospital and Harvard Medical School Boston, Massachusetts
| |
Collapse
|
35
|
Podoltsev NA, Wang R, Shallis RM, Stempel JM, Di M, Neparidze N, Zeidan AM, Huntington SF, Giri S, Hull SC, Gore SD, Ma X. Statin use, survival and incidence of thrombosis among older patients with polycythemia vera and essential thrombocythemia. Cancer Med 2023; 12:18889-18900. [PMID: 37702132 PMCID: PMC10557879 DOI: 10.1002/cam4.6528] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Polycythemia vera (PV) and essential thrombocythemia (ET) are linked to increased risk of cardiovascular morbidity and mortality. In addition to the reduction in of arterial thrombotic events, statins may prevent venous thrombosis including among patients with cancer. As previous registry- and claims-based studies revealed that the use of statins may improve the survival of patients with various malignancies we evaluated their impact on outcomes of older adults with PV and ET. METHODS We identified 4010 older adults (aged 66-99 years at diagnosis) with PV (n = 1809) and ET (n = 2201) in a population-based cohort study using the Surveillance, Epidemiology, and End Results-Medicare database with median follow-up of 3.92 (interquartile range: 2.58-5.75) years. Propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) approaches were utilized to assess potential association between statins and overall survival. Multivariable competing risk models with death as a competing risk were used to evaluate possible relationship between statins and the incidence of thrombosis. RESULTS 55.8% of the patients used statins within the first year after PV/ET diagnosis, and statin use was associated with a 22% reduction in all-cause mortality (PSM: hazard ratio [HR] = 0.78, 95% confidence interval [CI]: 0.63-0.98, p = 0.03; IPTW: HR = 0.79, 95% CI: 0.64-0.97, p = 0.03). Statins also reduced the risk of thrombosis in this patient population (PSM: HR = 0.63, 95% CI: 0.51-0.78, p < 0.01; IPTW: HR = 0.57, 95% CI: 0.49-0.66, p < 0.01) as well as in PV and ET subgroups. CONCLUSIONS These findings suggest that it may be important to incorporate statins into the therapeutic strategy for older adults with PV and ET.
Collapse
Affiliation(s)
- Nikolai A. Podoltsev
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Rong Wang
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
| | - Rory M. Shallis
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Jessica M. Stempel
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Mengyang Di
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Natalia Neparidze
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Scott F. Huntington
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
| | - Smith Giri
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Present address:
Division of Hematology and OncologyUniversity of Alabama School of MedicineBirminghamAlabamaUSA
| | - Sarah C. Hull
- Section of Cardiology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Steven D. Gore
- Section of Hematology, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Present address:
Investigational Drug Branch, Cancer Therapy Evaluation ProgramNational Cancer InstituteBethesdaMarylandUSA
| | - Xiaomei Ma
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) CenterYale UniversityNew HavenConnecticutUSA
- Department of Chronic Disease EpidemiologyYale School of Public HealthNew HavenConnecticutUSA
| |
Collapse
|
36
|
Hasselbalch HC, Junker P, Skov V, Kjær L, Knudsen TA, Larsen MK, Holmström MO, Andersen MH, Jensen C, Karsdal MA, Willumsen N. Revisiting Circulating Extracellular Matrix Fragments as Disease Markers in Myelofibrosis and Related Neoplasms. Cancers (Basel) 2023; 15:4323. [PMID: 37686599 PMCID: PMC10486581 DOI: 10.3390/cancers15174323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023] Open
Abstract
Philadelphia chromosome-negative chronic myeloproliferative neoplasms (MPNs) arise due to acquired somatic driver mutations in stem cells and develop over 10-30 years from the earliest cancer stages (essential thrombocythemia, polycythemia vera) towards the advanced myelofibrosis stage with bone marrow failure. The JAK2V617F mutation is the most prevalent driver mutation. Chronic inflammation is considered to be a major pathogenetic player, both as a trigger of MPN development and as a driver of disease progression. Chronic inflammation in MPNs is characterized by persistent connective tissue remodeling, which leads to organ dysfunction and ultimately, organ failure, due to excessive accumulation of extracellular matrix (ECM). Considering that MPNs are acquired clonal stem cell diseases developing in an inflammatory microenvironment in which the hematopoietic cell populations are progressively replaced by stromal proliferation-"a wound that never heals"-we herein aim to provide a comprehensive review of previous promising research in the field of circulating ECM fragments in the diagnosis, treatment and monitoring of MPNs. We address the rationales and highlight new perspectives for the use of circulating ECM protein fragments as biologically plausible, noninvasive disease markers in the management of MPNs.
Collapse
Affiliation(s)
- Hans Carl Hasselbalch
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Peter Junker
- Department of Rheumatology, Odense University Hospital, 5000 Odense, Denmark;
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Trine A. Knudsen
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Morten Kranker Larsen
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Herlev Hospital, 2730 Herlev, Denmark; (M.O.H.); (M.H.A.)
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Herlev Hospital, 2730 Herlev, Denmark; (M.O.H.); (M.H.A.)
| | - Christina Jensen
- Nordic Bioscience A/S, 2730 Herlev, Denmark; (C.J.); (M.A.K.); (N.W.)
| | - Morten A. Karsdal
- Nordic Bioscience A/S, 2730 Herlev, Denmark; (C.J.); (M.A.K.); (N.W.)
| | | |
Collapse
|
37
|
Verstovsek S, Mesa R, Gupta V, Lavie D, Dubruille V, Cambier N, Platzbecker U, Hus M, Xicoy B, Oh ST, Kiladjian JJ, Vannucchi AM, Gerds A, Egyed M, Mayer J, Sacha T, Kawashima J, Morris M, Huang M, Harrison C. Momelotinib long-term safety and survival in myelofibrosis: integrated analysis of phase 3 randomized controlled trials. Blood Adv 2023; 7:3582-3591. [PMID: 37042865 PMCID: PMC10368854 DOI: 10.1182/bloodadvances.2022009311] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/08/2023] [Accepted: 03/06/2023] [Indexed: 04/13/2023] Open
Abstract
Momelotinib is the first inhibitor of Janus kinase 1 (JAK1) and JAK2 shown to also inhibit activin A receptor type 1 (ACVR1), a key regulator of iron homeostasis, and has demonstrated improvements in splenomegaly, constitutional symptoms, and anemia in myelofibrosis (MF). This long-term analysis pooled data from 3 randomized phase 3 studies of momelotinib (MOMENTUM, SIMPLIFY-1, and SIMPLIFY-2), representing MF disease from early (JAK inhibitor-naive) to late (JAK inhibitor-experienced) stages. Patients in the control arms (danazol in MOMENTUM, ruxolitinib in SIMPLIFY-1, and best available therapy in SIMPLIFY-2) could cross over to receive momelotinib at the end of the 24-week randomized period, and all patients could continue momelotinib treatment after the completion of these studies via an extended access protocol (XAP). Across these studies, 725 patients with MF received momelotinib; 12% remained on therapy for ≥5 years, with a median treatment exposure of 11.3 months (range, 0.1-90.4 months). The most common nonhematologic treatment-emergent adverse event (AE) occurring in ≥20% of patients was diarrhea (any grade, 27% and grade ≥3, 3%). Any-grade thrombocytopenia, anemia, and neutropenia occurred in 25%, 23%, and 7% of patients, respectively. The most common reason for momelotinib discontinuation was thrombocytopenia (4% discontinuation rate). The incidence of AEs of clinical importance (eg, infections, malignant transformation, peripheral neuropathy, and hemorrhage) did not increase over time. This analysis of one of the largest randomized trial databases for a JAK inhibitor to date in MF demonstrated a consistent safety profile of momelotinib without long-term or cumulative toxicity. These trials were registered at www.clinicaltrials.gov as: MOMENTUM (#NCT04173494), SIMPLIFY-1 (#NCT01969838), SIMPLIFY-2 (#NCT02101268), and XAP (#NCT03441113).
Collapse
Affiliation(s)
| | - Ruben Mesa
- UT Health San Antonio Cancer Center, San Antonio, TX
| | - Vikas Gupta
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - David Lavie
- Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Viviane Dubruille
- Centre Hospitalier Universitaire de Nantes (CHU de Nantes), Nantes, France
| | - Nathalie Cambier
- Service d’hématologie, Centre hospitalier régional universitaire de Lille (CHRU Lille), Lille, France
| | - Uwe Platzbecker
- Clinic of Hematology, Cellular Therapy, and Hemostaseology, University of Leipzig, Leipzig, Germany
| | - Marek Hus
- Uniwersytet Medyczny w Lublinie, Lublin, Poland
| | - Blanca Xicoy
- Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Stephen T. Oh
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jean-Jacques Kiladjian
- Université de Paris, Assistance Publique–Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM, Paris, France
| | - Alessandro M. Vannucchi
- Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, University of Florence, Careggi University Hospital, Florence, Italy
| | | | | | - Jiří Mayer
- Department of Internal Medicine, Haematology and Oncology, University Hospital Brno, Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tomasz Sacha
- Uniwersytet Jagielloński Collegium Medicum, Krakow, Poland
| | | | | | | | - Claire Harrison
- Guy's and St Thomas' National Health Services (NHS) Foundation Trust, London, United Kingdom
| |
Collapse
|
38
|
Leiva O, Jenkins A, Rosovsky RP, Leaf RK, Goodarzi K, Hobbs G. Risk Factors for Death or Cardiovascular Events after Acute Coronary Syndrome in Patients with Myeloproliferative Neoplasms. Hematol Rep 2023; 15:398-404. [PMID: 37367089 DOI: 10.3390/hematolrep15020040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/21/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Patients with myeloproliferative neoplasms (MPNs) are at increased risk of cardiovascular disease (CVD), including acute coronary syndrome (ACS). However, data on long-term outcomes of patients with MPN who have had ACS and risk factors for all-cause death or CV events post-ACS hospitalization are lacking. We conducted a single-center study of 41 consecutive patients with MPN with ACS hospitalization after MPN diagnosis. After a median follow-up of 80 months after ACS hospitalization, 31 (76%) experienced death or a CV event (myocardial infarction, ischemic stroke, or heart failure hospitalization). After multivariable Cox proportional hazards regression, index ACS within 12 months of MPN diagnosis (HR 3.84, 95% CI 1.44-10.19), WBC ≥ 20 K/µL (HR 9.10, 95% CI 2.71-30.52), JAK2 mutation (HR 3.71, 95% CI 1.22-11.22), and prior CVD (HR 2.60, 95% CI 1.12-6.08) were associated with increased death or CV events. Further studies are warranted to improve cardiovascular outcomes in this patient population.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiovascular Medicine, Department of Medicine, New York University Langone Health, New York, NY 10016, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Andrew Jenkins
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Rachel P Rosovsky
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Rebecca K Leaf
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Katayoon Goodarzi
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Gabriela Hobbs
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
39
|
Leiva O, Hobbs G. Author's reply. J Cardiol 2023; 81:587-588. [PMID: 36681318 DOI: 10.1016/j.jjcc.2023.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023]
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Gabriela Hobbs
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Predictors of increased risk of adverse cardiovascular outcomes among patients with myeloproliferative neoplasms and atrial fibrillation. J Cardiol 2023; 81:260-267. [PMID: 36384716 DOI: 10.1016/j.jjcc.2022.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Patients with myeloproliferative neoplasms (MPNs), essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF), have increased risk of cardiovascular (CV) disease. Atrial fibrillation (AF) is associated with adverse CV outcomes including arterial thrombosis, heart failure (HF), and CV death and coexists with MPN. Traditional risk scores (CHA2DS2-VASC and HAS-BLED) for estimating risks/benefits of anticoagulation to prevent thrombotic events in AF do not include MPN status. Therefore, we aimed to investigate CV outcomes in patients with MPN and AF and evaluate the predictive ability of traditional risk scores. METHODS We conducted a single-center, retrospective cohort study of patients with MPN and AF. Primary outcome was composite of CV death and arterial thromboembolism; secondary outcomes were bleeding requiring emergency department visit or hospitalization, hospitalization for HF, and all-cause death. Multivariable competing-risk and Cox proportional hazards regression models were used to estimate risk of outcomes. Receiver operating characteristic (ROC) curve used to evaluate predictive ability of CHA2DS2-VASC and HAS-BLED of composite outcome and bleeding, respectively. RESULTS A total 142 patients was included (62 ET, 54 PV, 26 MF). Composite outcome, bleeding, HF hospitalization and all-cause death occurred in 39 %, 30 %, 34 %, and 48 %, of patients respectively. After multivariable modeling, MF was associated with increased risk of composite outcome (SHR 2.70, 95 % CI 1.38-5.27) and all-cause mortality (HR 9.77, 95 % CI 4.88-19.54) but not bleeding (SHR 1.19, 95 % CI 0.51-2.80) or HF admissions (SHR 0.57, 95 % CI 0.19-1.72). CHA2DS2-VASC and HAS-BLED were poor predictors of composite outcome (C-statistic 0.52, 95 % CI 0.43-0.62) and bleeding (C-statistic 0.49, 95 % CI 0.40-0.58), respectively. CONCLUSION In patients with MPN and AF, MF is associated with increased risk of CV death and arterial thrombosis and traditional risk scores do not accurately predict outcomes in this patient population. Further investigation is needed to refine risk scores in this patient population.
Collapse
|
41
|
Singh S, Singh J, Mehta A, Sharma R, Joshi K, Jain K, Paul D, Oberoi G, Jindal N, Dhillon B, Narang V. Distinctive Attributes of Indian Patients With Classical BCR::ABL1 Negative Myeloproliferative Neoplasms: Unified Clinical and Laboratory Data. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:360-369.e1. [PMID: 36849307 DOI: 10.1016/j.clml.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/29/2023]
Abstract
INTRODUCTION We report one of the largest single center data from a mixed referral setting in India describing baseline characteristics and outcomes of patients with classical BCR::ABL1 negative myeloproliferative neoplasms (MPNs). MATERIALS AND METHODS Patients diagnosed from June 2019 to 2022 were included. Workup and treatment was as per current guidelines. RESULTS Diagnosis comprised polycythemia vera (PV) in 51(49%), ET in 33(31.7%) and prefibrotic primary myelofibrosis (MF) pre fibrotic myelofibrosis (prePMF) and myelofibrosis in 10(9.6%) patients each. Median age at diagnosis was 52 years for PV and ET, 65.5 for MF and 79 years for prePMF. Diagnosis was incidental in 63(56.7%) and after thrombosis in 8(7.2%) patients. Baseline next generation sequencing (NGS) was available for 63(60.5%) patients. Driver mutations in PV: JAK2 in 80.3%; in ET: JAK2 in 41%, CALR in 26%, MPL in 2.9%; in prePMF JAK2 in 70%, CALR in 20%, MPL in 10%, and in MF: JAK2 in 10%, MPL in 30% and CALR in 40%. Seven novel mutations were detected of which 5 were potentially pathogenic on computational analysis. After median follow up of 30 months, 2 patients had disease transformation and none had new episodes of thrombosis. Ten patients died, most commonly with cardiovascular events(n = 5,50%). Median overall survival was not reached. Mean OS time was 10.19 years(95%CI, 8.6 to 11.74) and mean time to transformation was 12.2 years(95% CI,11.8 to 12.6). CONCLUSION Our data indicates comparatively indolent presentation of MPNs in India with younger age and lower risk of thrombosis. Further follow up will enable correlation with molecular data and guide modification of age based risk stratification models.
Collapse
Affiliation(s)
- Suvir Singh
- Department of Clinical Haematology and Stem Cell Transplantation, Dayanand Medical College and Hospital, Ludhiana, India.
| | - Jagdeep Singh
- Department of Medical Oncology, Dayanand Medical College and Hospital, Ludhiana, India
| | - Arpan Mehta
- Neuberg Supratech Reference Laboratories, Ahmedabad, India
| | - Rintu Sharma
- Department of Clinical Haematology and Stem Cell Transplantation, Dayanand Medical College and Hospital, Ludhiana, India
| | - Kaveri Joshi
- Department of Clinical Haematology and Stem Cell Transplantation, Dayanand Medical College and Hospital, Ludhiana, India
| | - Kunal Jain
- Department of Medical Oncology, Dayanand Medical College and Hospital, Ludhiana, India
| | - Davinder Paul
- Department of Medical Oncology, Dayanand Medical College and Hospital, Ludhiana, India
| | - Gurleen Oberoi
- Department of Hematopathology, All India Institute of Medical Sciences, New Delhi, India
| | - Nandita Jindal
- Department of Molecular Genetics, Dayanand Medical College and Hospital, Ludhiana, India
| | - Barjinderjit Dhillon
- Department of Molecular Genetics, Dayanand Medical College and Hospital, Ludhiana, India
| | - Vikram Narang
- Department of Pathology, Dayanand Medical College and Hospital, Ludhiana, India
| |
Collapse
|
42
|
Găman MA, Kipkorir V, Srichawla BS, Dhali A, Găman AM, Diaconu CC. Primary Arterial Hypertension and Drug-Induced Hypertension in Philadelphia-Negative Classical Myeloproliferative Neoplasms: A Systematic Review. Biomedicines 2023; 11:388. [PMID: 36830925 PMCID: PMC9952891 DOI: 10.3390/biomedicines11020388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
The impact of primary arterial hypertension (HTN) in myeloproliferative neoplasms (MPNs) remains unclear, with scant literature available, mostly focusing on cardiovascular risk factors as a singular entity or on organ-specific HTN. Furthermore, available studies reporting findings on drug-induced HTN in MPNs report varying and contradictory findings. In consideration of the above, this study set out to systematically review the available literature and shed light on the occurrence of HTN in MPNs, its association with thrombosis, as well as the drugs used in MPN management that could increase blood pressure. The literature search yielded 598 potentially relevant records of which 315 remained after the duplicates (n = 283) were removed. After we screened the titles and the abstracts of these publications, we removed irrelevant papers (n = 228) and evaluated the full texts of 87 papers. Furthermore, 13 records did not meet the inclusion criteria and were excluded from the systematic review. Finally, a total of 74 manuscripts were entered into the qualitative synthesis and included in the present systematic review. Our systematic review highlights that HTN is the most common comorbidity encountered in MPNs, with an impact on both the occurrence of thrombosis and survival. Moreover, drug-induced HTN remains a challenge in the management of MPNs. Further research should investigate the characteristics of patients with MPNs and HTN, as well as clarify the contribution of HTN to the development of thrombotic complications, survival and management in MPNs. In addition, the relationship between clonal hematopoiesis of indeterminate potential, HTN, cardiovascular disease and MPNs requires examination in upcoming assessments.
Collapse
Affiliation(s)
- Mihnea-Alexandru Găman
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Vincent Kipkorir
- Department of Human Anatomy and Physiology, University of Nairobi, Nairobi 30197, Kenya
| | | | - Arkadeep Dhali
- Department of Internal Medicine, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, UK
| | - Amelia Maria Găman
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Clinic of Hematology, Filantropia City Hospital, 200143 Craiova, Romania
| | - Camelia Cristina Diaconu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| |
Collapse
|
43
|
Baragetti A, Suurmond J, Marques PE, Tavares LP. Editorial: Rising stars in inflammation 2021. Front Immunol 2023; 14:1193694. [PMID: 37207223 PMCID: PMC10189773 DOI: 10.3389/fimmu.2023.1193694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Affiliation(s)
- A. Baragetti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy
- *Correspondence: A. Baragetti,
| | - J. Suurmond
- Department of Rheumatology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - P. E. Marques
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - L. P. Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
44
|
Pemmaraju N, Harrison C, Gupta V, Verstovsek S, Scott B, Oh ST, Palandri F, Al‐Ali HK, Sobas M, McMullin MF, Mesa R, Buckley S, Roman‐Torres K, Vannucchi A, Yacoub A. Risk-adjusted safety analysis of the oral JAK2/IRAK1 inhibitor pacritinib in patients with myelofibrosis. EJHAEM 2022; 3:1346-1351. [PMID: 36467816 PMCID: PMC9713029 DOI: 10.1002/jha2.591] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/21/2022] [Indexed: 06/17/2023]
Abstract
The safety profile of the novel oral JAK2/IRAK1 inhibitor pacritinib in patients with cytopenic myelofibrosis was described in the Phase 2 PAC203 and Phase 3 PERSIST-2 studies. To account for longer treatment durations on the pacritinib arms compared to best available therapy (BAT), we present a risk-adjusted safety analysis of event rates accounting for different time on treatment. While the rate of overall events was higher on pacritinib compared to BAT, the rate of fatal events was lower, and there was no excess in bleeding, cardiac events, secondary malignancy, or thrombosis on pacritinib, including in patients with severe thrombocytopenia.
Collapse
Affiliation(s)
- Naveen Pemmaraju
- The University of Texas, MD Anderson Cancer CenterHoustonTexasUSA
| | | | - Vikas Gupta
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioCanada
| | - Srdan Verstovsek
- The University of Texas, MD Anderson Cancer CenterHoustonTexasUSA
| | - Bart Scott
- Fred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Stephen T. Oh
- Washington University School of MedicineSt LouisMissouriUSA
| | - Francesca Palandri
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaIstituto di Ematologia “Seràgnoli”BolognaItaly
| | | | - Marta Sobas
- Department of Hematology, Blood Neoplasms and Bone Marrow TransplantationWroclaw Medical UniversityWrocławPoland
| | | | - Ruben Mesa
- UT Health San Antonio MD Anderson Cancer CenterSan AntonioTexasUSA
| | | | | | | | | |
Collapse
|
45
|
Mulas O, Mola B, Madeddu C, Caocci G, Macciò A, Nasa GL. Prognostic Role of Cell Blood Count in Chronic Myeloid Neoplasm and Acute Myeloid Leukemia and Its Possible Implications in Hematopoietic Stem Cell Transplantation. Diagnostics (Basel) 2022; 12:2493. [PMID: 36292182 PMCID: PMC9600993 DOI: 10.3390/diagnostics12102493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/01/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Numerous prognostic indexes have been developed in hematological diseases based on patient characteristics and genetic or molecular assessment. However, less attention was paid to more accessible parameters, such as neutrophils, lymphocytes, monocytes, and platelet counts. Although many studies have defined the role of neutrophil-to-lymphocyte or platelet-to-lymphocyte in lymphoid malignancies, few applications exist for myeloid neoplasm or hematopoietic stem cell transplantation procedures. In this review, we synthesized literature data on the prognostic value of count blood cells in myeloid malignancies and hematopoietic stem cell transplantation in the context of classical prognostic factors and clinical outcomes.
Collapse
Affiliation(s)
- Olga Mulas
- Hematology Unit, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
| | - Brunella Mola
- Hematology Unit, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
| | - Giovanni Caocci
- Hematology Unit, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
| | - Antonio Macciò
- Department of Gynecologic Oncology, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
- Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Giorgio La Nasa
- Hematology Unit, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
| |
Collapse
|
46
|
Bhatnagar R, Dixit NM, Yang EH, Sallam T. Cancer therapy's impact on lipid metabolism: Mechanisms and future avenues. Front Cardiovasc Med 2022; 9:925816. [PMID: 36017084 PMCID: PMC9396263 DOI: 10.3389/fcvm.2022.925816] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerotic cardiovascular disease is a growing threat among cancer patients. Not surprisingly, cancer-targeting therapies have been linked to metabolic dysregulation including changes in local and systemic lipid metabolism. Thus, tumor development and cancer therapeutics are intimately linked to cholesterol metabolism and may be a driver of increased cardiovascular morbidity and mortality in this population. Chemotherapeutic agents affect lipid metabolism through diverse mechanisms. In this review, we highlight the mechanistic and clinical evidence linking commonly used cytotoxic therapies with cholesterol metabolism and potential opportunities to limit atherosclerotic risk in this patient population. Better understanding of the link between atherosclerosis, cancer therapy, and cholesterol metabolism may inform optimal lipid therapy for cancer patients and mitigate cardiovascular disease burden.
Collapse
Affiliation(s)
- Roshni Bhatnagar
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Neal M. Dixit
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Eric H. Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tamer Sallam
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|