1
|
Qamar U, Agarwal S, Khawandanah M, Pandey M, AlJuhaishi T, Asad ZUA. Cardiovascular disease-related mortality trends among adults with multiple myeloma in the United States from 1999 to 2023. Leuk Res 2025; 154:107713. [PMID: 40344949 DOI: 10.1016/j.leukres.2025.107713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/08/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Affiliation(s)
- Usama Qamar
- Department of Internal Medicine, Mayo Clinic, Rochester, USA.
| | | | - Mohamad Khawandanah
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Manu Pandey
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Taha AlJuhaishi
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Zain Ul Abideen Asad
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| |
Collapse
|
2
|
Pan Y, Chen B, Xie J, Chen D, Cai Y, Zhao D, Cao Y, Lian F, Yan X. Lentinan alleviates angiotensin II-induced myocardial remodeling through LMP7-SOCS3 signaling. Int J Biol Macromol 2025; 308:142146. [PMID: 40101827 DOI: 10.1016/j.ijbiomac.2025.142146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Myocardial remodeling is a major pathological mechanism causing heart failure. As a critical negative modulator of cardiac remodeling, suppressor of cytokine signaling 3 (SOCS3) is regulated by immunoproteasome subunit large multifunctional peptidase 7 (LMP7). Lentinan (LNT), a β-polysaccharide extracted from Lentinus edodes, has anti-inflammatory and antioxidant properties. However, the role and molecular mechanisms of LNT in angiotensin II (Ang II)-triggered myocardial remodeling are unclear. Myocardial remodeling was established using Ang II infusion (1000 or 200 ng/kg/min) for 2 weeks. Cardiomyocytes and cardiac fibroblasts were triggered by Ang II. LNT was administered daily by oral gavage to mice starting 1 day before Ang II or saline treatment. Here, we found that LNT supplementation dose-dependently ameliorated Ang II-triggered myocardial dysfunction and remodeling (hypertrophy, fibrosis, inflammation, and superoxide production). Mechanistically, LNT suppressed SOCS3 protein degradation by downregulating immunoproteasome LMP7 activity and expression, thereby inactivating downstream signaling, such as STAT3, ERK, AKT, NF-κB, and TGF-β. Conversely, SOCS3 knockdown significantly blocked the protective effect of LNT on myocardial remodeling in Ang II-infused mice. Together, our findings suggest that LNT may be a new therapeutic approach for myocardial remodeling and heart failure.
Collapse
Affiliation(s)
- Yu Pan
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Bingqi Chen
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Jiawen Xie
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Danni Chen
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Yuwei Cai
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Denghui Zhao
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Yifei Cao
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Fuzhi Lian
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China; Engineering Research Center of Mobile Health Management System, Ministry of Education, Hangzhou, China
| | - Xiao Yan
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China.
| |
Collapse
|
3
|
Gertz MA. Moving the Therapeutic Needle in Immunoglobulin Light Chain and TTR Cardiac Amyloidosis. Am J Hematol 2025; 100:900-902. [PMID: 39953923 DOI: 10.1002/ajh.27642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Affiliation(s)
- Morie A Gertz
- Dvision of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
4
|
Hinterndorfer M, Spiteri VA, Ciulli A, Winter GE. Targeted protein degradation for cancer therapy. Nat Rev Cancer 2025:10.1038/s41568-025-00817-8. [PMID: 40281114 DOI: 10.1038/s41568-025-00817-8] [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] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
Targeted protein degradation (TPD) aims at reprogramming the target specificity of the ubiquitin-proteasome system, the major cellular protein disposal machinery, to induce selective ubiquitination and degradation of therapeutically relevant proteins. Since its conception over 20 years ago, TPD has gained a lot of attention mainly due to improvements in the design of bifunctional proteolysis targeting chimeras (PROTACs) and understanding the mechanisms underlying molecular glue degraders. Today, PROTACs are on the verge of a first clinical approval and recent structural and mechanistic insights combined with technological leaps promise to unlock the rational design of protein degraders, following the lead of lenalidomide and related clinically approved analogues. At the same time, the TPD universe is expanding at a record speed with the discovery of novel modalities beyond molecular glue degraders and PROTACs. Here we review the recent progress in the field, focusing on newly discovered degrader modalities, the current state of clinical degrader candidates for cancer therapy and upcoming design approaches.
Collapse
Affiliation(s)
- Matthias Hinterndorfer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Valentina A Spiteri
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, UK
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, UK.
| | - Georg E Winter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
5
|
Morimoto S, Ichihara A. Impact of blood pressure variability on the prediction of cancer therapy-related cardiovascular toxicity in patients with multiple myeloma. Hypertens Res 2025:10.1038/s41440-025-02215-x. [PMID: 40240871 DOI: 10.1038/s41440-025-02215-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025]
Affiliation(s)
- Satoshi Morimoto
- Department of Medicine, Tokyo Women's Medical University Adachi Medical Center, Tokyo, Japan.
- Division of Hormonal Medicine and Bioregulatory Science, Department of Medicine, Tokyo Women's Medical University, Tokyo, Japan.
| | - Atsuhiro Ichihara
- Division of Hormonal Medicine and Bioregulatory Science, Department of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
6
|
Ding Y, Chen R, Zhou J, Bao Y, Meng N, Zheng X, Yang S, Lu J, Jiang Z, Liu Y, Xie C, Lu L, Lu W. All-stage targeted nanodiscs for glioma treatment by inducing cuproptosis and apoptosis of cancer cells and cancer stem cells. Asian J Pharm Sci 2025; 20:101010. [PMID: 40182135 PMCID: PMC11964743 DOI: 10.1016/j.ajps.2024.101010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/21/2024] [Accepted: 11/05/2024] [Indexed: 04/05/2025] Open
Abstract
There remain several intractable challenges for chemotherapy in glioma treatment, including the blood-brain barrier (BBB), blood-brain tumor barrier (BBTB), and tumor heterogeneity caused by cancer stem cells (CSCs), which are resistant to conventional chemotherapy. Here, we established a nano strategy to kill glioma cells and CSCs, combining carfilzomib and bis(diethyldithiocarbamate)copper. The synergistic drug combination disturbed cell protein metabolism at different stages and induced apoptosis and cuproptosis. The Y-shaped targeting ligand pHA-VAP-modified nanodiscs were designed to help the chemotherapeutic agents cross the BBB/BBTB and finally accumulate in tumor site. This all-stage targeting and all-stage treatment nanomedicine significantly prolonged the survival in glioma-bearing mice and might inspire the rational design of advanced drug delivery platforms.
Collapse
Affiliation(s)
- Yuan Ding
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Ruohan Chen
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jianfen Zhou
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Yanning Bao
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Nana Meng
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Xudong Zheng
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Shengmin Yang
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jiasheng Lu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Zhixuan Jiang
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Yu Liu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Cao Xie
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Weiyue Lu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China
| |
Collapse
|
7
|
Fanelli E, Picca G, Airale L, Astarita A, Mingrone G, Catarinella C, Votta S, Colomba A, Cesareo M, Leone D, Paladino A, Rabbia F, Bringhen S, Gay F, Veglio F, Milan A, Vallelonga F. Blood pressure variability as predictor of cancer therapy-related cardiovascular toxicity in patients with Multiple Myeloma. Hypertens Res 2025; 48:1554-1563. [PMID: 39843857 DOI: 10.1038/s41440-024-02084-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/14/2024] [Accepted: 12/21/2024] [Indexed: 01/24/2025]
Abstract
Blood pressure (BP) variability (BPV) is an independent predictor of cardiovascular (CV) events. The role of BPV in defining risk of cancer therapy-related cardiovascular toxicity (CTR-CVT) is currently unknown. The aims of this study were: (i) to evaluate BPV in a population of patients with Multiple Myeloma, undergoing proteasome inhibitors therapy; (ii) to assess the predictive value of BPV for CTR-CVT; (iii) to analyze clusters of subjects based on BPV. One hundred twenty-four patients underwent a baseline evaluation, including Ambulatory Blood Pressure Monitoring (ABPM), PWV, and Echocardiography. BPV was assessed through ABPM-based standard deviation (SD), weighted standard deviation (wSD), coefficient of variation (CoV), average real variability (ARV), and variability independent of the mean (VIM). Individuals who developed CTR-CVT had a higher baseline BPV. Furthermore, night-time BPV was associated with CTR-CVT, independently of age, smoking, BP, diabetes, dyslipidemia, and kidney function (night-time systolic CoV: adjusted OR 1.09 [1.01-1.21]; night-time systolic VIM: adjusted OR 1.18 [1.01-1.39]). Cut-offs for these BPV parameters were identified as predictors of CTR-CVT occurrence: 10.5 for night-time systolic CoV; 7.8 and 6.4 for systolic and diastolic night-time VIM. Clustering analysis identified subgroups of subjects characterized by the highest BPV, who had a greater prevalence of events, but no differences in other CV risk determinants. Short-term BPV is an independent predictor of CTR-CVT. BPV may enhance the precision of risk stratification in cancer patients, enabling identification of individuals at higher risk who would not be recognized, if traditional prognostic indicators were the sole applied criteria. On the left panel in the figure, the distribution of blood pressure variability (BPV) in the population according to cancer therapy-related cardiovascular toxicity occurrence; in the central panel, association of blood pressure variability with events and cutoffs values; in the right panel, clustering analysis results based on BPV levels. Histogram and radar plot represent events and BPV indexes distribution in the three clusters, respectively. ARV, average real variability; BPV, Blood Pressure Variability; CTR-CVT, cancer therapy-related cardiovascular toxicity; CoV, coefficient of variation; DBP, Diastolic blood pressure; SBP, Systolic blood pressure; SD, standard deviation; VIM, variability independent of the mean; wSD, weighted standard deviation.
Collapse
Affiliation(s)
- Elvira Fanelli
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy.
- Emergency Medicine Unit, Ospedale San Giovanni Bosco, Turin, Italy.
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Giulia Picca
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Lorenzo Airale
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Astarita
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giulia Mingrone
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Cinzia Catarinella
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Simona Votta
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Colomba
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Marco Cesareo
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Dario Leone
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Arianna Paladino
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Franco Rabbia
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Sara Bringhen
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Francesca Gay
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, AOU Città della Salute e della Scienza di Torino, Turin, Italy
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Franco Veglio
- Division of Internal Medicine, Hypertension Unit, A.O.U. Città della Salute e della Scienza di Torino, Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alberto Milan
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fabrizio Vallelonga
- Division of Internal Medicine, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
8
|
Sawant P, Mathew A, Bensalel J, Gallego-Delgado J, Mandal P. Cancer Drug Bortezomib, a Proteasomal Inhibitor, Triggers Cytotoxicity in Microvascular Endothelial Cells via Multi-Organelle Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645304. [PMID: 40196627 PMCID: PMC11974872 DOI: 10.1101/2025.03.25.645304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Proteasomes maintain cellular homeostasis by degrading abnormal proteins, while cancer cells exploit them for survival, becoming a key chemotherapeutic target. Bortezomib (BTZ), a reversible proteasomal inhibitor, is a front-line treatment for multiple myeloma, mantle cell lymphoma, and non-small cell lung cancer. However, its efficacy is limited by severe side effects, including neurotoxicity and cardiovascular distress, with its toxicity mechanisms largely unexplored. Here, we discover that Bortezomib (BTZ), is cytotoxic to non-cancerous cells distinctly from Carfilzomib (CFZ), the second-line irreversible PI. BTZ or CFZ is administered intravenously, impacting blood vessel (vascular) endothelial cells. We used human pulmonary microvascular endothelial cells (HPMECs) to demonstrate that BTZ but not CFZ elicits endoplasmic reticulum (ER) stress, mitochondrial membrane compromise, mitochondrial reactive oxygen species (ROS) accumulation, and Caspase (CASP)9 activation (mediator of Intrinsic apoptosis) within fifteen hours of treatment. By twenty-four hours, BTZ-treated cells display cleavage of CASP8 (mediator of extrinsic apoptosis), activation of CASP3 (terminal executioner of apoptosis), cell-death and vascular barrier loss. Pan-caspase inhibitor zVAD significantly rescues BTZ-treated cells from cytotoxicity. Both BTZ and CFZ effectively kill MM cells. These findings reveal novel insights into fundamental signaling of regular cells where reversible inhibition of the proteasome dictates a unique cascade of stress distinct from irreversible inhibition. These harmful effects of BTZ emphasize the need to re- evaluate its use as a frontline chemotherapy for MM. Highlights Reversible proteasomal inhibitor Bortezomib is cytotoxic to non-cancerous, microvascular endothelial cellsIn endothelial cells, Bortezomib, but not irreversible inhibitor Carfilzomib, activates temporal cascade of caspases (Caspase-9, Caspase-8, Caspase-3) triggering apoptosisCaspase activation results from ER stress (via the IRE1α-CHOP) pathway and mitochondrial stress (ROS accumulation) independently from contribution from extrinsic signal via TNFBortezomib-dependent cytotoxicity compromises endothelial barrier potential.
Collapse
|
9
|
Pan QM, Bi FF, Jing ZH, Cao M, Cui C, Liu F, Jin L, Yi-Jie H, Tian H, Yu T, Yun W, Shan HL, Zhou YH. A New target of ischemic ventricular arrhythmias-ITFG2. Eur J Pharmacol 2025; 991:177301. [PMID: 39864577 DOI: 10.1016/j.ejphar.2025.177301] [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: 10/26/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
ITFG2 is an intracellular protein known to modulate the immune response of T-cells. Our previous investigation revealed that ITFG2 specifically targets ATP5b to regulate ATP energy metabolism and maintain mitochondrial function, thereby protecting the heart from ischemic injury. However, the role of ITFG2 in ischemic ventricular arrhythmias and its underlying mechanisms have not been previously reported. In this study, we found ITFG2 overexpression, induced by an adeno-associated virus serotype 9 vector, partially reduced the incidence of ischemic ventricular arrhythmias and shortened the duration of ventricular arrhythmias in mice after myocardial infarction. Conversely, shRNA-mediated knockdown of endogenous ITFG2 aggravated ischemic ventricular arrhythmias. ITFG2 overexpression also shortened the prolonged QRS complex and increased the epicardial conduction velocity in MI mice. Additionally, the hearts from ITFG2 overexpression mice exhibited a higher maximal upstroke velocity at phase 0 of transmembrane action potential compared to MI mice. Patch-clamp analyses demonstrated a 50% increase in the peak current of voltage-dependent Na+ channel by ITFG2 overexpression in isolated ventricular cardiomyocytes post MI. In cultured neonatal mouse cardiomyocytes under hypoxic conditions, ITFG2 up-regulated Nav1.5 protein expression by inhibiting its ubiquitination. Co-immunoprecipitation experiments showed that ITFG2 reduces the binding affinity between NEDD4-2 and Nav1.5, thereby inhibiting Nav1.5 ubiquitination. Taken together, our data highlight the critical role of ITFG2 in reducing susceptibility to ischemic ventricular arrhythmias by down-regulating Nav1.5 ubiquitination. These findings suggest that ITFG2 may serve as a novel target for treating ischemic ventricular arrhythmias.
Collapse
Affiliation(s)
- Qing-Ming Pan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Fang-Fang Bi
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Ze-Hong Jing
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Miao Cao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Chen Cui
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Fu Liu
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College, Xiamen Medical College, Xiamen, Fujian, 361023, PR China
| | - Li Jin
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College, Xiamen Medical College, Xiamen, Fujian, 361023, PR China; Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - He Yi-Jie
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College, Xiamen Medical College, Xiamen, Fujian, 361023, PR China; Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Hua Tian
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College, Xiamen Medical College, Xiamen, Fujian, 361023, PR China
| | - Tong Yu
- Hanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, PR China
| | - Wu Yun
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Hong-Li Shan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China; Hanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, PR China.
| | - Yu-Hong Zhou
- Department of Basic Medicine, Institute of Respiratory Diseases Xiamen Medical College, Xiamen Medical College, Xiamen, Fujian, 361023, PR China; State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, Heilongjiang, 150081, PR China.
| |
Collapse
|
10
|
Ahammed MS, Wang X. Promoting proteostasis by cAMP/PKA and cGMP/PKG. Trends Mol Med 2025; 31:224-239. [PMID: 39477759 PMCID: PMC11908951 DOI: 10.1016/j.molmed.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 11/06/2024]
Abstract
Proteasome functional insufficiency (PFI) is implicated in neurodegeneration and heart failure, where aberrant protein aggregation is common and impairs the ubiquitin (Ub)-proteasome system (UPS), exacerbating increased proteotoxic stress (IPTS) and creating a vicious circle. Breaking this circle represents a key to treating these diseases. Protein kinase (PK)-A and PKG can activate the proteasome and promote proteasomal degradation of misfolded proteins. PKA does so by phosphorylating Ser14-RPN6/PSMD11, but how PKG activates the proteasome remains unknown. Emerging evidence supports a strategy to treat diseases with IPTS by augmenting cAMP/PKA and cGMP/PKG. Conceivably, targeted activation of PKA and PKG at proteasome nanodomains would minimize the undesired effects from their actions on other targets. In this review, we discuss PKA and PKG regulation of proteostasis via the UPS.
Collapse
Affiliation(s)
- Md Salim Ahammed
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA.
| |
Collapse
|
11
|
Basin S, Cezar M, Fraix A, Pace N, Filippetti L, Schulmann S, Selton-Suty C, Huttin O, Chabot F, Chaouat A, Valentin S. A longitudinal study of right ventricular function of patients with multiple myeloma treated with carfilzomib. ERJ Open Res 2025; 11:00861-2024. [PMID: 40129546 PMCID: PMC11931522 DOI: 10.1183/23120541.00861-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/06/2024] [Indexed: 03/26/2025] Open
Abstract
A particular focus on right ventricular function should be suggested in patients during treatment with carfilzomib due to potential RV side-effects. TAPSE follow-up appears to be an interesting tool for close monitoring of these patients. https://bit.ly/4dTedb9.
Collapse
Affiliation(s)
- Sarah Basin
- Centre Hospitalier Universitaire de Nancy, Département de Pneumologie, Vandœuvre-lès-Nancy, France
| | - Mathilde Cezar
- Centre Hospitalier Universitaire de Nancy, Département de Pneumologie, Vandœuvre-lès-Nancy, France
| | - Antoine Fraix
- Service de Cardiologie, Institut Lorrain du Cœur et des Vaisseaux, Centre Hospitalier Universitaire de Nancy, Nancy, France
| | - Nathalie Pace
- Service de Cardiologie, Institut Lorrain du Cœur et des Vaisseaux, Centre Hospitalier Universitaire de Nancy, Nancy, France
| | - Laura Filippetti
- Service de Cardiologie, Institut Lorrain du Cœur et des Vaisseaux, Centre Hospitalier Universitaire de Nancy, Nancy, France
| | | | - Christine Selton-Suty
- Service de Cardiologie, Institut Lorrain du Cœur et des Vaisseaux, Centre Hospitalier Universitaire de Nancy, Nancy, France
| | - Olivier Huttin
- Service de Cardiologie, Institut Lorrain du Cœur et des Vaisseaux, Centre Hospitalier Universitaire de Nancy, Nancy, France
| | - François Chabot
- Centre Hospitalier Universitaire de Nancy, Département de Pneumologie, Vandœuvre-lès-Nancy, France
- INSERM, UMR_S 1116, Université de Lorraine, Nancy, France
| | - Ari Chaouat
- Centre Hospitalier Universitaire de Nancy, Département de Pneumologie, Vandœuvre-lès-Nancy, France
- INSERM, UMR_S 1116, Université de Lorraine, Nancy, France
| | - Simon Valentin
- Centre Hospitalier Universitaire de Nancy, Département de Pneumologie, Vandœuvre-lès-Nancy, France
- INSERM U1254, IADI, Université de Lorraine, Nancy, France
| |
Collapse
|
12
|
Zhou Y, Chen Y, Xu M, Zhang Y, Wan X, Xia Y, Wang H, Zeng H. The effect of proteasome in heart transplantation: From mechanisms to therapeutic potential. Life Sci 2025; 364:123446. [PMID: 39920983 DOI: 10.1016/j.lfs.2025.123446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Heart transplantation is a critical treatment for end-stage heart failure. However, its clinical efficacy is hindered by some challenges, such as ischemia-reperfusion injury (IRI) and post-transplant rejection. These complications significantly contribute to graft dysfunction and compromise patient survival. Emerging evidence underscores the involvement of proteasome in the pathophysiology of both IRI and post-transplant rejection. Proteasome inhibition has demonstrated potential in attenuating IRI by limiting oxidative damage and apoptosis while also mitigating rejection through the regulation of adaptive and innate immune responses. Recent advances in the development of proteasome inhibitors, particularly in optimizing specificity and minimizing adverse effects, have further strengthened their prospects for clinical application. This review focuses on the roles of the proteasome and its inhibitors in heart transplantation, with an emphasis on their mechanisms and therapeutic applications in managing IRI and rejection.
Collapse
Affiliation(s)
- Ye Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030, Hubei, China
| | - Yu Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Mengyao Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ying Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030, Hubei, China
| | - Xiaoning Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yudong Xia
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongjie Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030, Hubei, China.
| | - Hesong Zeng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030, Hubei, China.
| |
Collapse
|
13
|
Jarnes JR, Pillai NR, Ahmed A, Shrestha S, Stark M, Whitley CB. Dose-intensive therapy (DIT) for infantile Pompe disease: A pilot study. Mol Genet Metab Rep 2025; 42:101179. [PMID: 39802096 PMCID: PMC11720876 DOI: 10.1016/j.ymgmr.2024.101179] [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: 10/21/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
Background The current standard of care for infantile-onset Pompe disease (IOPD), a severe form of acid α-glucosidase enzyme activity deficiency is: (1) detection by newborn screening, (2) early initiation of intravenous enzyme replacement therapy (ERT) using recombinant human acid alpha-glucosidase (rhGAA), with higher doses of rhGAA increasingly used to improve clinical outcomes, and (3) immune tolerization induction (ITI) using to prevent anti-rhGAA antibody formation, with methotrexate (MTX), rituximab, and IVIG used for patients who are cross-reactive immunologic material negative (CRIM-) and monotherapy with MTX used in patients who are cross-reactive immunologic material positive (CRIM+). Objectives/methods A pilot study evaluates a dose-intensive therapy (DIT) using high-dose ERT (40 mg/kg/week) and more frequent exposure to ERT (i.e., 3 times weekly administration) to mitigate anti-rhGAA antibody formation, as an alternative to the standard therapeutic approach for IOPD. Results In the first patient, DIT resulted in rapid normalization of the following: (1) bi-ventricular hypertrophy, (2) urine HEX-4, (3) CK, (4) liver transaminases. At 7 years of age, the patient continues the DIT regimen. To date, all pediatric developmental milestones have been met on time, anti-rhGAA antibodies have been negative and the patient is able to attend school and maintain normal activities of daily living. Conclusions Over a 7-year period, DIT for CRIM-positive IOPD was well tolerated in the first patient treated. Excellent clinical outcomes were achieved, and anti-rhGAA antibodies levels were consistently undetectable. Assessments of more patients, that includes patients with CRIM-, as well as CRIM+ IOPD, will determine if this approach consistently achieves improved clinical outcomes and immune tolerization.
Collapse
Affiliation(s)
- Jeanine R. Jarnes
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Advanced Therapies Program, University of Minnesota, Fairview, Minneapolis, MN, USA
- Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Nishitha R. Pillai
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Advanced Therapies Program, University of Minnesota, Fairview, Minneapolis, MN, USA
| | - Alia Ahmed
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Advanced Therapies Program, University of Minnesota, Fairview, Minneapolis, MN, USA
| | - Sofia Shrestha
- Advanced Therapies Program, University of Minnesota, Fairview, Minneapolis, MN, USA
- M Health Fairview Masonic Children's Hospital, Minneapolis, MN, USA
| | - Molly Stark
- M Health Fairview Masonic Children's Hospital, Minneapolis, MN, USA
| | - Chester B. Whitley
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Advanced Therapies Program, University of Minnesota, Fairview, Minneapolis, MN, USA
- Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
14
|
Rasooly D, Giambartolomei C, Peloso GM, Dashti H, Ferolito BR, Golden D, Horimoto ARVR, Pietzner M, Farber-Eger EH, Wells QS, Bini G, Proietti G, Tartaglia GG, Kosik NM, Wilson PWF, Phillips LS, Munroe PB, Petersen SE, Cho K, Gaziano JM, Leach AR, Whittaker J, Langenberg C, Aung N, Sun YV, Pereira AC, Casas JP, Joseph J. Large-scale multi-omics identifies drug targets for heart failure with reduced and preserved ejection fraction. NATURE CARDIOVASCULAR RESEARCH 2025; 4:293-311. [PMID: 39915329 DOI: 10.1038/s44161-025-00609-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/07/2025] [Indexed: 03/19/2025]
Abstract
Heart failure (HF) has limited therapeutic options. In this study, we differentiated the pathophysiological underpinnings of the HF subtypes-HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF)-and uncovered subtype-specific therapeutic strategies. We investigated the causal roles of the human proteome and transcriptome using Mendelian randomization on more than 420,000 participants from the Million Veteran Program (27,799 HFrEF and 27,579 HFpEF cases). We created therapeutic target profiles covering efficacy, safety, novelty, druggability and mechanism of action. We replicated findings on more than 175,000 participants of diverse ancestries. We identified 70 HFrEF and 10 HFpEF targets, of which 58 were not previously reported; notably, the HFrEF and HFpEF targets are non-overlapping, suggesting the need for subtype-specific therapies. We classified 14 previously unclassified HF loci as HFrEF. We substantiated the role of ubiquitin-proteasome system, small ubiquitin-related modifier pathway, inflammation and mitochondrial metabolism in HFrEF. Among druggable genes, IL6R, ADM and EDNRA emerged as potential HFrEF targets, and LPA emerged as a potential target for both subtypes.
Collapse
Affiliation(s)
- Danielle Rasooly
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA.
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Claudia Giambartolomei
- Integrative Data Analysis Unit, Health Data Science Centre, Human Technopole, Milan, Italy
| | - Gina M Peloso
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Hesam Dashti
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Brian R Ferolito
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA
| | - Daniel Golden
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea R V R Horimoto
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maik Pietzner
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Addenbrookes Hospital, IMS, Cambridge, UK
- Computational Medicine, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Eric H Farber-Eger
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quinn Stanton Wells
- Departments of Medicine (Cardiology), Biomedical Informatics and Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Giorgio Bini
- Istituto Italiano di Tecnologia, CHT@Erzelli, Genova, Italy
- Dipartimento di Fisica Via Dodecaneso, Genova, Italy
| | | | - Gian Gaetano Tartaglia
- Istituto Italiano di Tecnologia, CHT@Erzelli, Genova, Italy
- ICREA - Institució Catalana de Recerca I Estudis Avançats, Barcelona, Spain
| | - Nicole M Kosik
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA
| | - Peter W F Wilson
- Atlanta VA Health Care System, Decatur, GA, USA
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lawrence S Phillips
- Atlanta VA Health Care System, Decatur, GA, USA
- Division of Endocrinology and Metabolism, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Steffen E Petersen
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Kelly Cho
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - J Michael Gaziano
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew R Leach
- Department of Chemical Biology, European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| | - John Whittaker
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Addenbrookes Hospital, IMS, Cambridge, UK
- Computational Medicine, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Nay Aung
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Yan V Sun
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexandre C Pereira
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo, São Paulo, Brazil
| | - Juan P Casas
- Million Veteran Program (MVP) Coordinating Center, Veterans Affairs Healthcare System, Boston, MA, USA
| | - Jacob Joseph
- Cardiology Section, VA Providence Healthcare System, Providence, RI, USA.
- Warren Alpert School of Medicine, Brown University, Providence, RI, USA.
| |
Collapse
|
15
|
Kelly C, Kiltschewskij DJ, Leong AJW, Haw TJ, Croft AJ, Balachandran L, Chen D, Bond DR, Lee HJ, Cairns MJ, Sverdlov AL, Ngo DTM. Identifying common pathways for doxorubicin and carfilzomib-induced cardiotoxicities: transcriptomic and epigenetic profiling. Sci Rep 2025; 15:4395. [PMID: 39910168 PMCID: PMC11799237 DOI: 10.1038/s41598-025-87442-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
Cancer therapy-related cardiovascular toxicity (CTR-CVT) is now recognised as one of the leading causes of long-term morbidity and mortality in cancer patients. To date, potential overlapping cardiotoxicity mechanism(s) across different chemotherapeutic classes have not been elucidated. Doxorubicin, an anthracycline, and Carfilzomib, a proteasome inhibitor, are both known to cause heart failure in some patients. Given this common cardiotoxic effect of these chemotherapies, we aimed to investigate differential and common mechanism(s) associated with Doxorubicin and Carfilzomib-induced cardiac dysfunction. Primary human cardiomyocyte-like cells (HCM-ls) were treated with 1 µM of either Doxorubicin or Carfilzomib for 72 h. Both Doxorubicin and Carfilzomib induced a significant reduction in HCM cell viability and cell damage. DNA methylation analysis performed using MethylationEPIC array showed distinct and common changes induced by Doxorubicin and Carfilzomib (10,270 or approximately 12.9% of the DMPs for either treatment overlapped). RNA-seq analyses identified 5,643 differentially expressed genes (DEGs) that were commonly dysregulated for both treatments. Pathway analysis revealed that the PI3K-Akt signalling pathway was the most significantly enriched pathway with common DEGs, shared between Doxorubicin and Carfilzomib. We identified that there are shared cardiotoxicity mechanisms for Doxorubicin and Carfilzomib pathways that can be potential therapeutic targets for treatments across 2 classes of anti-cancer agents.
Collapse
Affiliation(s)
- Conagh Kelly
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
| | - Dylan J Kiltschewskij
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Angeline J W Leong
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Tatt Jhong Haw
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Amanda J Croft
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Lohis Balachandran
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Dongqing Chen
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
| | - Danielle R Bond
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Heather J Lee
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Aaron L Sverdlov
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia.
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia.
- Cardiovascular Department, John Hunter Hospital, New Lambton Heights, NSW, Australia.
| | - Doan T M Ngo
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia.
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia.
| |
Collapse
|
16
|
Bloom MW, Vo JB, Rodgers JE, Ferrari AM, Nohria A, Deswal A, Cheng RK, Kittleson MM, Upshaw JN, Palaskas N, Blaes A, Brown SA, Ky B, Lenihan D, Maurer MS, Fadol A, Skurka K, Cambareri C, Chauhan C, Barac A. Cardio-Oncology and Heart Failure: a Scientific Statement From the Heart Failure Society of America. J Card Fail 2025; 31:415-455. [PMID: 39419165 DOI: 10.1016/j.cardfail.2024.08.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Heart failure and cancer remain 2 of the leading causes of morbidity and mortality, and the 2 disease entities are linked in a complex manner. Patients with cancer are at increased risk of cardiovascular complications related to the cancer therapies. The presence of cardiomyopathy or heart failure in a patient with new cancer diagnosis portends a high risk for adverse oncology and cardiovascular outcomes. With the rapid growth of cancer therapies, many of which interfere with cardiovascular homeostasis, heart failure practitioners need to be familiar with prevention, risk stratification, diagnosis, and management strategies in cardio-oncology. This Heart Failure Society of America statement addresses the complexities of heart failure care among patients with active cancer diagnoses and cancer survivors. Risk stratification, monitoring and management of cardiotoxicity are presented across stages A through D heart failure, with focused discussion on heart failure with preserved ejection fraction and special populations, such as survivors of childhood and young-adulthood cancers. We provide an overview of the shared risk factors between cancer and heart failure, highlighting heart failure as a form of cardiotoxicity associated with many different cancer therapeutics. Finally, we discuss disparities in the care of patients with cancer and cardiac disease and present a framework for a multidisciplinary-team approach and critical collaboration among heart failure, oncology, palliative care, pharmacy, and nursing teams in the management of these complex patients.
Collapse
Affiliation(s)
| | - Jacqueline B Vo
- Radiation Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Bethesda, MD
| | - Jo E Rodgers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC
| | - Alana M Ferrari
- Division of Hematology/ Oncology, University of Virginia Health, Charlottesville, VA
| | - Anju Nohria
- Cardio-Oncology Program, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard K Cheng
- Division of Cardiology, University of Washington, Seattle, WA
| | - Michelle M Kittleson
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Nicolas Palaskas
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anne Blaes
- Division of Hematology/Oncology/Transplantation, University of Minnesota, Minneapolis, MN
| | - Sherry-Ann Brown
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI; Research Collaborator, Mayo Clinic, Rochester, MN
| | - Bonnie Ky
- Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Thalheimer Center for Cardio-Oncology, Abramson Cancer Center and Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel Lenihan
- Saint Francis Healthcare, Cape Girardeau, MO and the International Cardio-Oncology Society, Tampa, FL
| | - Mathew S Maurer
- Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | | | | | - Christine Cambareri
- Clinical Oncology Pharmacist, Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA
| | | | - Ana Barac
- Department of Cardiology, Inova Schar Heart and Vascular, Inova Schar Cancer, Falls Church, VA
| |
Collapse
|
17
|
Sayegh H, Zagouras A, Neal JW, Witteles RM, Zhu H, Waliany S. Classes of Antineoplastic Agents Associated with Increased Risk of Cancer Therapy-associated Hypertension and Management Strategies. Cardiol Clin 2025; 43:31-42. [PMID: 39551560 DOI: 10.1016/j.ccl.2024.08.006] [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] [Indexed: 11/19/2024]
Abstract
Hypertension (HTN) has been found to be the most common comorbidity in patients with cancer. In addition to increased prevalence of baseline HTN, patients with cancer may be at increased risk of HTN as a short-term or long-term adverse event from cancer therapy. Different classes of cancer therapies have been implicated in the development of HTN, including inhibitors of vascular endothelial growth factor (VEGF), Bruton tyrosine kinase inhibitors, proteasome inhibitors, androgen deprivation therapy, and others. While some of these drugs may lead to increases in blood pressure through on-target effects (eg, with VEGF inhibition), others may be associated with HTN from off-target mechanisms that are not always well understood.
Collapse
Affiliation(s)
- Hoda Sayegh
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexia Zagouras
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joel W Neal
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cancer Institute, Stanford, CA, USA
| | - Ronald M Witteles
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Han Zhu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Sarah Waliany
- Massachusetts General Hospital Cancer Center, Boston, MA, USA; Dana-Farber Cancer Institute, Boston, MA 02114, USA.
| |
Collapse
|
18
|
Chang YC, Peng CY, Chi KY, Song J, Chang Y, Chiang CH, Gao W, Chiang CH. Cardiovascular outcomes and mortality in diabetic multiple myeloma patients initiated on proteasome inhibitors according to prior use of glucagon-like peptide 1 agonists. Eur J Prev Cardiol 2025:zwaf017. [PMID: 39878555 DOI: 10.1093/eurjpc/zwaf017] [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] [Received: 09/04/2024] [Revised: 11/02/2024] [Accepted: 12/31/2024] [Indexed: 01/31/2025]
Abstract
Lay Summary
Glucagon-like peptide 1 agonists may reduce the risk of major adverse cardiovascular events, heart failure, and all-cause mortality among patients with multiple myeloma and Type 2 diabetes mellitus receiving proteasome inhibitors. The use of glucagon-like peptide 1 agonist is associated with a reduction in cardiovascular events and mortality among patients with multiple myeloma and Type 2 diabetes mellitus receiving proteasome inhibitors. The use of glucagon-like peptide 1 agonist is not associated with an increased risk of adverse events among patients with multiple myeloma and Type 2 diabetes mellitus receiving proteasome inhibitors.
Collapse
Affiliation(s)
- Yu-Cheng Chang
- Department of Medicine, Danbury Hospital, Danbury, CT, USA
| | - Chun-Yu Peng
- Department of Medicine, Danbury Hospital, Danbury, CT, USA
| | - Kuan-Yu Chi
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Junmin Song
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yu Chang
- Section of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cho-Hung Chiang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wenli Gao
- Department of Medicine, Danbury Hospital, Danbury, CT, USA
| | - Cho-Han Chiang
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, 330 Mt Auburn St, Cambridge, MA 02138, USA
| |
Collapse
|
19
|
Tong J, Senechal I, Ramalingam S, Lyon AR. Risk Assessment Prior to Cardiotoxic Anticancer Therapies in 7 Steps. Br J Hosp Med (Lond) 2025; 86:1-21. [PMID: 39862029 DOI: 10.12968/hmed.2024.0632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Abstract
The burdens of cardiovascular (CV) diseases and cardiotoxic side effects of cancer treatment in oncology patients are increasing in parallel. The European Society of Cardiology (ESC) 2022 Cardio-Oncology guidelines recommend the use of standardized risk stratification tools to determine the risk of cardiotoxicity associated with different anticancer treatment modalities and the severity of their complications. The use of the Heart Failure Association-International Cardio-Oncology Society (HFA-ICOS) is essential for assessing risk prior to starting cancer treatment, and validation of these methods has been performed in patients receiving anthracyclines, human epidermal receptor 2 (HER2)-targeted therapies and breakpoint cluster region-abelson oncogene locus (BCR-ABL) inhibitors. The benefits of performing baseline CV risk assessment and stratification include early recognition of cardiotoxicities, personalisation of cancer treatment and monitoring strategies, and allocation of cardioprotection to those at the highest risk. This review summarizes the key points of risk stratification in these patients. The steps include identifying the target population, assessing nonmodifiable and modifiable CV risk factors, reviewing previous oncologic therapies and CV histories, and performing baseline investigations. In summary, this review aims to provide general physicians with a simple 7-step guide that will help steer and navigate them through cardiac risk evaluation of potentially cardiotoxic oncologic treatment strategies.
Collapse
Affiliation(s)
- Jieli Tong
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
- Department of Cardiology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Isabelle Senechal
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| | | | - Alexander R Lyon
- Cardio-Oncology Centre of Excellence, Royal Brompton Hospital, London, UK
| |
Collapse
|
20
|
Alshahrani AA, Kontopantelis E, Morgan C, Ravindrarajah R, Martin GP, Mamas MA. Cardiovascular diseases in patients with cancer: A comprehensive review of epidemiological trends, cardiac complications, and prognostic implications. Chin Med J (Engl) 2025; 138:143-154. [PMID: 39719690 PMCID: PMC11745857 DOI: 10.1097/cm9.0000000000003419] [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: 05/28/2024] [Indexed: 12/26/2024] Open
Abstract
ABSTRACT This article provides an overview of the current evidence on the epidemiology, overlapping risk factors, and pathophysiology of cardiovascular disease (CVD) in patients with cancer. It explores the cardiotoxic effects of anticancer therapy and their impact on prognosis. Although cancer survival rates have improved over the last two decades, the risk of CVD has risen over time in patients with cancer. CVD and cancer share similar risk factors and a common pathophysiology involving inflammation. Many chemotherapeutic agents used to treat cancer are associated with cardiovascular complications (such as heart failure, myocardial infarction, and thrombosis). Current evidence indicates a significant burden of CVD in patients with cancer, particularly in the first year following cancer diagnosis, with elevated risk persisting beyond this period. This short- and long-term risk of CVD may vary depending on the cancer type and treatment regimen. Early identification of potential cardiovascular risk in patients with cancer, can lead to more favorable clinical and survival outcomes. Given the acute and long-term consequences, patients with cancer require increased cardiovascular care and lifestyle optimization. This article offers valuable insights into the cardiovascular burden and needs of patients with cancer. It is intended for a general medical research readership interested in the intersection of cardiology and oncology.
Collapse
Affiliation(s)
- Ali A. Alshahrani
- Faculty of Biology, Medicine and Health, School of Health Sciences, University of Manchester, Manchester M13 9QQ, United Kingdom
- Department of Invasive Cardiovascular Technology, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh 3129, Saudi Arabia
| | - Evangelos Kontopantelis
- Faculty of Biology, Medicine and Health, School of Health Sciences, University of Manchester, Manchester M13 9QQ, United Kingdom
| | - Catharine Morgan
- Faculty of Biology, Medicine and Health, School of Health Sciences, University of Manchester, Manchester M13 9QQ, United Kingdom
| | - Rathi Ravindrarajah
- Faculty of Biology, Medicine and Health, School of Health Sciences, University of Manchester, Manchester M13 9QQ, United Kingdom
| | - Glen P. Martin
- Faculty of Biology, Medicine and Health, School of Health Sciences, University of Manchester, Manchester M13 9QQ, United Kingdom
| | - Mamas A. Mamas
- Keele Cardiovascular Research Group, Institute for Prognosis Research, University of Keele, Keele ST5 5BG, United Kingdom
| |
Collapse
|
21
|
Bernasconi R, Kuster GM. Non-coding RNAs and their potential exploitation in cancer therapy-related cardiotoxicity. Br J Pharmacol 2025; 182:296-315. [PMID: 38802331 DOI: 10.1111/bph.16416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/28/2024] [Accepted: 03/26/2024] [Indexed: 05/29/2024] Open
Abstract
Life expectancy in cancer patients has been extended in recent years, thanks to major breakthroughs in therapeutic developments. However, this also unmasked an increased incidence of cardiovascular diseases in cancer survivors, which is in part attributable to cancer therapy-related cardiovascular toxicity. Non-coding RNAs (ncRNAs) have received much appreciation due to their impact on gene expression. NcRNAs, which include microRNAs, long ncRNAs and circular RNAs, are non-protein-coding transcripts that are involved in the regulation of various biological processes, hence shaping cell identity and behaviour. They have also been implicated in disease development, including cardiovascular diseases, cancer and, more recently, cancer therapy-associated cardiotoxicity. This review outlines key features of cancer therapy-associated cardiotoxicity, what is known about the roles of ncRNAs in these processes and how ncRNAs could be exploited as therapeutic targets for cardioprotection. LINKED ARTICLES: This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
Affiliation(s)
- Riccardo Bernasconi
- Myocardial Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Gabriela M Kuster
- Myocardial Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Cardiology, University Heart Center Basel, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
22
|
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
|
23
|
Molnár AÁ, Birgés K, Surman A, Merkely B. The Complex Connection Between Myocardial Dysfunction and Cancer Beyond Cardiotoxicity: Shared Risk Factors and Common Molecular Pathways. Int J Mol Sci 2024; 25:13185. [PMID: 39684895 DOI: 10.3390/ijms252313185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Cardiovascular diseases and cancer represent the largest disease burden worldwide. Previously, these two conditions were considered independent, except in terms of cardiotoxicity, which links cancer treatment to subsequent cardiovascular issues. However, recent studies suggest that there are further connections between cancer and heart disease beyond cardiotoxicity. It has been revealed that myocardial dysfunction may promote carcinogenesis, indicating that additional common pathophysiological mechanisms might be involved in the relationship between cardiology and oncology, rather than simply a connection through cardiotoxic effects. These mechanisms may include shared risk factors and common molecular pathways, such as persistent inflammation and neurohormonal activation. This review explores the connection between myocardial dysfunction and cancer, emphasizing their shared risk factors, similar biological mechanisms, and causative factors like cardiotoxicity, along with their clinical implications.
Collapse
Affiliation(s)
| | - Kristóf Birgés
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| | - Adrienn Surman
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary
| |
Collapse
|
24
|
Sato AY, Cregor M, McAndrews K, Schurman CA, Schaible E, Shutter J, Vyas P, Adhikari B, Willis MS, Boerma M, Alliston T, Bellido T. Pharmacologic or genetic interference with atrogene signaling protects against glucocorticoid-induced musculoskeletal and cardiac disease. JCI Insight 2024; 9:e182664. [PMID: 39405125 PMCID: PMC11601705 DOI: 10.1172/jci.insight.182664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/18/2024] [Indexed: 11/09/2024] Open
Abstract
Despite their beneficial actions as immunosuppressants, glucocorticoids (GC) have devastating effects on the musculoskeletal and cardiac systems, as long-term treated patients exhibit high incidence of falls, bone fractures, and cardiovascular events. Herein, we show that GC upregulate simultaneously in bone, skeletal muscle, and the heart the expression of E3 ubiquitin ligases (atrogenes), known to stimulate the proteasomal degradation of proteins. Activation of vitamin D receptor (VDR) signaling with the VDR ligands calcitriol or eldecalcitol prevented GC-induced atrogene upregulation in vivo and ex vivo in bone/muscle organ cultures and preserved tissue structure/mass and function of the 3 tissues in vivo. Direct pharmacologic inhibition of the proteasome with carfilzomib also conferred musculoskeletal protection. Genetic loss of the atrogene MuRF1-mediated protein ubiquitination in ΔRING mice afforded temporary or sustained protection from GC excess in bone or skeletal and heart muscle. We concluded that the atrogene pathway downstream of MuRF1 underlies GC action in bone, muscle, and the heart, and it can be pharmacologically or genetically targeted to confer protection against the damaging actions of GC simultaneously in the 3 tissues.
Collapse
Affiliation(s)
- Amy Y. Sato
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Meloney Cregor
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kevin McAndrews
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Charles A. Schurman
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Eric Schaible
- Advanced Light Source, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Jennifer Shutter
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Punit Vyas
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bhawana Adhikari
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Monte S. Willis
- Allegheny Health Network, Pathology and Laboratory Medicine Institute, Pittsburgh, Pennsylvania, USA
- Quest Diagnostics Inc., NE Regional Core Lab, Clifton, New Jersey, USA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Marjan Boerma
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, USA
| | - Teresita Bellido
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
- Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
25
|
Raisi-Estabragh Z, Murphy AC, Ramalingam S, Scherrer-Crosbie M, Lopez-Fernandez T, Reynolds KL, Aznar M, Lin AE, Libby P, Cordoba R, Bredsen-Masley C, Wechalekar A, Apperley J, Cheng RK, Manisty CH. Cardiovascular Considerations Before Cancer Therapy: Gaps in Evidence and JACC: CardioOncology Expert Panel Recommendations. JACC CardioOncol 2024; 6:631-654. [PMID: 39479317 PMCID: PMC11520216 DOI: 10.1016/j.jaccao.2024.07.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 11/02/2024] Open
Abstract
Baseline cardiovascular assessment before the initiation of potentially cardiotoxic cancer therapies is a key component of cardio-oncology, aiming to reduce cardiovascular complications and morbidity in patients and survivors. Recent clinical practice guidelines provide both general and cancer therapy-specific recommendations for baseline cardiovascular toxicity risk assessment and management, including the use of dedicated risk scores, cardiovascular imaging, and biomarker testing. However, the value of such interventions in altering disease trajectories has not been established, with many recommendations based on expert opinion or Level of Evidence: C, studies with a potential for high risk of bias. Advances in understanding underlying mechanisms of cardiotoxicity and the increased availability of genetic and immunologic profiling present new opportunities for personalized risk assessment. This paper evaluates the existing evidence on cardiovascular care of cancer patients before cardiotoxic cancer therapy and highlights gaps in evidence and priorities for future research.
Collapse
Affiliation(s)
- Zahra Raisi-Estabragh
- William Harvey Research Institute, National Institute for Health and Care Research Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kindgom
| | | | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Marielle Scherrer-Crosbie
- Division of Cardiology, The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa Lopez-Fernandez
- Hospital Universitario La Paz, Instituto de Investigación La Paz-IdiPaz, Madrid, Spain
- Hospital Universitario Quiron Pozuelo, Madrid, Spain
| | - Kerry L. Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marianne Aznar
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Amy E. Lin
- Department of Medicine, Division of Cardiology, Section of Cardio-Oncology and Immunology, University of California-San Francisco, San Francisco, California, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Raul Cordoba
- Fundacion Jimenez Diaz University Hospital, Health Research Institute Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Christine Bredsen-Masley
- Division of Hematology/Oncology, Department of Medicine, St. Michael’s Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Ashu Wechalekar
- Department of Haematology, University College London Hospitals NHS Trust, London, United Kingdom
| | - Jane Apperley
- Department of Clinical Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Richard K. Cheng
- Division of Cardiology, University of Washington Medical Center, Seattle, Washington, USA
| | - Charlotte H. Manisty
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kindgom
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| |
Collapse
|
26
|
Price LC, Lyon A, Boyd K. Reversible carfilzomib-induced pulmonary arterial hypertension: don't take your eyes off the ball! Eur Respir J 2024; 64:2401087. [PMID: 39389614 DOI: 10.1183/13993003.01087-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/30/2024] [Indexed: 10/12/2024]
Affiliation(s)
- Laura C Price
- National Pulmonary Hypertension Service, Royal Brompton Hospital, London, UK
- NHLI Imperial College London, London, UK
| | - Alex Lyon
- Cardiology, Royal Brompton Hospital, London, UK
| | - Kevin Boyd
- Haematology, Royal Marsden Hospital, London, UK
| |
Collapse
|
27
|
Gao F, Xu T, Zang F, Luo Y, Pan D. Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms, Clinical Management and Innovative Treatment. Drug Des Devel Ther 2024; 18:4089-4116. [PMID: 39286288 PMCID: PMC11404500 DOI: 10.2147/dddt.s469331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/05/2024] [Indexed: 09/19/2024] Open
Abstract
With the continuous refinement of therapeutic measures, the survival rate of tumor patients has been improving year by year, while cardiovascular complications related to cancer therapy have become increasingly prominent. Exploring the mechanism and prevention strategy of cancer therapy-related cardiovascular toxicity (CTR-CVT) remains one of the research hotspots in the field of Cardio-Oncology in recent years. Cardiotoxicity of anticancer drugs involves heart failure, myocarditis, hypertension, arrhythmias and vascular toxicity, mechanistically related to vascular endothelial dysfunction, ferroptosis, mitochondrial dysfunction and oxidative stress. To address the cardiotoxicity induced by different anticancer drugs, various therapeutic measures have been put in place, such as reducing the accumulation of anticancer drugs, shifting to drugs with less cardiotoxicity, using cardioprotective drugs, and early detection. Due to the very limited treatments available to ameliorate anticancer drugs-induced cardiotoxicity, a few innovations are being shifted from animal studies to human studies. Examples include mitochondrial transplantation. Mitochondrial transplantation has been proven to be effective in in vivo and in vitro experiments. Several recent studies have demonstrated that intercellular mitochondrial transfer can ameliorate doxorubicin(DOX)-induced cardiotoxicity, laying the foundation for innovative therapies in anticancer drugs-induced cardiotoxicity. In this review, we will discuss the current status of anticancer drugs-induced cardiotoxicity in terms of the pathogenesis and treatment, with a focus on mitochondrial transplantation, and we hope that this review will bring some inspiration to you.
Collapse
Affiliation(s)
- Feiyu Gao
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Tao Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Fangnan Zang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| |
Collapse
|
28
|
Wilcox NS, Amit U, Reibel JB, Berlin E, Howell K, Ky B. Cardiovascular disease and cancer: shared risk factors and mechanisms. Nat Rev Cardiol 2024; 21:617-631. [PMID: 38600368 PMCID: PMC11324377 DOI: 10.1038/s41569-024-01017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Cardiovascular disease (CVD) and cancer are among the leading causes of morbidity and mortality globally, and these conditions are increasingly recognized to be fundamentally interconnected. In this Review, we present the current epidemiological data for each of the modifiable risk factors shared by the two diseases, including hypertension, hyperlipidaemia, diabetes mellitus, obesity, smoking, diet, physical activity and the social determinants of health. We then review the epidemiological data demonstrating the increased risk of CVD in patients with cancer, as well as the increased risk of cancer in patients with CVD. We also discuss the shared mechanisms implicated in the development of these conditions, highlighting their inherent bidirectional relationship. We conclude with a perspective on future research directions for the field of cardio-oncology to advance the care of patients with CVD and cancer.
Collapse
Affiliation(s)
- Nicholas S Wilcox
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Uri Amit
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob B Reibel
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eva Berlin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kendyl Howell
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Delimpasi S, Dimopoulos MA, Straub J, Symeonidis A, Pour L, Hájek R, Touzeau C, Bhanderi VK, Berdeja JG, Pavlíček P, Matous JV, Robak PJ, Suryanarayan K, Miller A, Villarreal M, Cherepanov D, Srimani JK, Yao H, Labotka R, Orlowski RZ. Ixazomib plus daratumumab and dexamethasone: Final analysis of a phase 2 study among patients with relapsed/refractory multiple myeloma. Am J Hematol 2024; 99:1746-1756. [PMID: 38856176 DOI: 10.1002/ajh.27382] [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: 11/08/2023] [Revised: 03/08/2024] [Accepted: 05/20/2024] [Indexed: 06/11/2024]
Abstract
Novel therapies have improved outcomes for multiple myeloma (MM) patients, but most ultimately relapse, making treatment decisions for relapsed/refractory MM (RRMM) patients increasingly challenging. We report the final analysis of a single-arm, phase 2 study evaluating the oral proteasome inhibitor (PI) ixazomib combined with daratumumab and dexamethasone (IDd; NCT03439293). Sixty-one RRMM patients (ixazomib/daratumumab-naïve; 1-3 prior therapies) were enrolled to receive IDd (28-day cycles) until disease progression/unacceptable toxicity. Median age was 69 years; 14.8% of patients had International Staging System stage III disease; 14.8% had received three prior therapies. Patients received a median of 16 cycles of IDd. In 59 response-evaluable patients, the overall response rate was 64.4%; the confirmed ≥very good partial response (VGPR) rate (primary endpoint) was 30.5%. Rates of ≥VGPR in patient subgroups were: high-risk cytogenetics (n = 15, 26.7%), expanded high-risk cytogenetics (n = 24, 29.2%), aged ≥75 years (n = 12, 16.7%), lenalidomide-refractory (n = 21, 28.6%), and prior PI/IMiD therapy (n = 58, 31.0%). With a median follow-up of 31.6 months, median progression-free survival was 16.8 months (95% confidence interval: 10.1-23.7). Grade ≥3 treatment-emergent adverse events (TEAEs) occurred in 54.1% of patients; 44.3% had serious TEAEs; TEAEs led to dose modifications/reductions/discontinuations in 62.3%/36.1%/16.4%. There were five on-study deaths. Any-grade and grade ≥3 peripheral neuropathy occurred in 18.0% and 1.6% of patients. Quality of life was generally maintained throughout treatment. IDd showed a positive risk-benefit profile in RRMM patients and was active in clinically relevant subgroups with no new safety signals.
Collapse
Affiliation(s)
- Sosana Delimpasi
- Department of Hematology and Bone Marrow Transplantation Unit, General Hospital Evangelismos, Athens, Greece
| | - Meletios A Dimopoulos
- Hematology & Medical Oncology, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Jan Straub
- Department of Internal Medicine - Hematology, University Hospital, Prague, Czech Republic
| | - Argiris Symeonidis
- Department of Hematology, University General Hospital of Patras, Patras, Greece
| | - Luděk Pour
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Roman Hájek
- Department of Haematooncology, University Hospital Ostrava and Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | | | | | | | - Petr Pavlíček
- Department of Internal Medicine and Hematology, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Jeffrey V Matous
- Colorado Blood Cancer Institute and Sarah Cannon Research Institute, Denver, Colorado, USA
| | - Pawel J Robak
- Department of Hematology, Medical University of Lodz and Copernicus Memorial Hospital, Lodz, Poland
| | - Kaveri Suryanarayan
- Clinical Research, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | - Alison Miller
- Statistics, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | - Miguel Villarreal
- Oncology, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | - Dasha Cherepanov
- Global Evidence and Outcomes (GEO), Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | - Jaydeep K Srimani
- Quantitative Clinical Pharmacology, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | - Huilan Yao
- Precision and Translational Medicine, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | - Richard Labotka
- Oncology Clinical Research, Takeda Development Center Americas, Inc. (TDCA), Lexington, Massachusetts, USA
| | - Robert Z Orlowski
- Departments of Lymphoma/Myeloma and Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
30
|
Bertolini D, Pizzi C, Donal E, Galli E. Cancer and Heart Failure: Dangerous Liaisons. J Cardiovasc Dev Dis 2024; 11:263. [PMID: 39330321 PMCID: PMC11432566 DOI: 10.3390/jcdd11090263] [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: 06/09/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
Cancer and heart failure (HF) are increasingly relevant worldwide, both from an epidemiologic and clinical point of view. This review aims to explore the relationship between cancer and HF by underscoring risk factors and disclosing the cardiotoxic effects of the current chemotherapy agents. We also deal with the current evidence on the diagnosis and management of HF related to cancer therapy. Finally, we will address the main gaps in knowledge and future perspectives in this field.
Collapse
Affiliation(s)
- Davide Bertolini
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences-DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
| | - Carmine Pizzi
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences-DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
| | - Erwan Donal
- Univ Rennes, CHU Rennes, Inserm, LTSI-UMR 1099, F-35000 Rennes, France
| | - Elena Galli
- Univ Rennes, CHU Rennes, Inserm, LTSI-UMR 1099, F-35000 Rennes, France
| |
Collapse
|
31
|
Logotheti S, Pavlopoulou A, Rudsari HK, Galow AM, Kafalı Y, Kyrodimos E, Giotakis AI, Marquardt S, Velalopoulou A, Verginadis II, Koumenis C, Stiewe T, Zoidakis J, Balasingham I, David R, Georgakilas AG. Intercellular pathways of cancer treatment-related cardiotoxicity and their therapeutic implications: the paradigm of radiotherapy. Pharmacol Ther 2024; 260:108670. [PMID: 38823489 DOI: 10.1016/j.pharmthera.2024.108670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/16/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Advances in cancer therapeutics have improved patient survival rates. However, cancer survivors may suffer from adverse events either at the time of therapy or later in life. Cardiovascular diseases (CVD) represent a clinically important, but mechanistically understudied complication, which interfere with the continuation of best-possible care, induce life-threatening risks, and/or lead to long-term morbidity. These concerns are exacerbated by the fact that targeted therapies and immunotherapies are frequently combined with radiotherapy, which induces durable inflammatory and immunogenic responses, thereby providing a fertile ground for the development of CVDs. Stressed and dying irradiated cells produce 'danger' signals including, but not limited to, major histocompatibility complexes, cell-adhesion molecules, proinflammatory cytokines, and damage-associated molecular patterns. These factors activate intercellular signaling pathways which have potentially detrimental effects on the heart tissue homeostasis. Herein, we present the clinical crosstalk between cancer and heart diseases, describe how it is potentiated by cancer therapies, and highlight the multifactorial nature of the underlying mechanisms. We particularly focus on radiotherapy, as a case known to often induce cardiovascular complications even decades after treatment. We provide evidence that the secretome of irradiated tumors entails factors that exert systemic, remote effects on the cardiac tissue, potentially predisposing it to CVDs. We suggest how diverse disciplines can utilize pertinent state-of-the-art methods in feasible experimental workflows, to shed light on the molecular mechanisms of radiotherapy-related cardiotoxicity at the organismal level and untangle the desirable immunogenic properties of cancer therapies from their detrimental effects on heart tissue. Results of such highly collaborative efforts hold promise to be translated to next-generation regimens that maximize tumor control, minimize cardiovascular complications, and support quality of life in cancer survivors.
Collapse
Affiliation(s)
- Stella Logotheti
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780, Athens, Greece; Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | | | - Anne-Marie Galow
- Institute for Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Yağmur Kafalı
- Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Efthymios Kyrodimos
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocrateion General Hospital Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Aris I Giotakis
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocrateion General Hospital Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Stephan Marquardt
- Institute of Translational Medicine for Health Care Systems, Medical School Berlin, Hochschule Für Gesundheit Und Medizin, 14197 Berlin, Germany
| | - Anastasia Velalopoulou
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, 35043 Marburg, Germany; German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany; Genomics Core Facility, Philipps-University, 35043 Marburg, Germany; Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Jerome Zoidakis
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece; Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, 18057 Rostock, Germany; Department of Life, Light & Matter, Interdisciplinary Faculty, Rostock University, 18059 Rostock, Germany
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780, Athens, Greece.
| |
Collapse
|
32
|
Mo CC, Richardson E, Calabretta E, Corrado F, Kocoglu MH, Baron RM, Connors JM, Iacobelli M, Wei LJ, Rapoport AP, Díaz-Ricart M, Moraleda JM, Carlo-Stella C, Richardson PG. Endothelial injury and dysfunction with emerging immunotherapies in multiple myeloma, the impact of COVID-19, and endothelial protection with a focus on the evolving role of defibrotide. Blood Rev 2024; 66:101218. [PMID: 38852017 DOI: 10.1016/j.blre.2024.101218] [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: 05/01/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Patients with multiple myeloma (MM) were among the groups impacted more severely by the COVID-19 pandemic, with higher rates of severe disease and COVID-19-related mortality. MM and COVID-19, plus post-acute sequelae of SARS-CoV-2 infection, are associated with endothelial dysfunction and injury, with overlapping inflammatory pathways and coagulopathies. Existing treatment options for MM, notably high-dose therapy with autologous stem cell transplantation and novel chimeric antigen receptor (CAR) T-cell therapies and bispecific T-cell engaging antibodies, are also associated with endothelial cell injury and mechanism-related toxicities. These pathologies include cytokine release syndrome (CRS) and neurotoxicity that may be exacerbated by underlying endotheliopathies. In the context of these overlapping risks, prophylaxis and treatment approaches mitigating the inflammatory and pro-coagulant effects of endothelial injury are important considerations for patient management, including cytokine receptor antagonists, thromboprophylaxis with low-molecular-weight heparin and direct oral anticoagulants, and direct endothelial protection with defibrotide in the appropriate clinical settings.
Collapse
Affiliation(s)
- Clifton C Mo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Edward Richardson
- Department of Medicine, Warren Alpert Medical School at Brown University, Providence, RI, USA
| | - Eleonora Calabretta
- Department of Biomedical Sciences, Humanitas University, and IRCCS Humanitas Research Hospital, Milan, Italy; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Francesco Corrado
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA; Department of Biomedical Sciences, Humanitas University, and IRCCS Humanitas Research Hospital, Milan, Italy; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Mehmet H Kocoglu
- Department of Medicine, University of Maryland School of Medicine, and Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Rebecca M Baron
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Lee-Jen Wei
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Aaron P Rapoport
- Department of Medicine, University of Maryland School of Medicine, and Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Maribel Díaz-Ricart
- Hematopathology, Pathology Department, CDB, Hospital Clinic, and IDIBAPS, Barcelona, Spain, and Barcelona Endothelium Team, Barcelona, Spain
| | - José M Moraleda
- Department of Medicine, Faculty of Medicine, Institute of Biomedical Research (IMIB-Pascual Parrilla), University of Murcia, Murcia, Spain
| | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University, and IRCCS Humanitas Research Hospital, Milan, Italy
| | - Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Kao TW, Huang TC, Liao CW, Shun CT, Tsai CH, Lin YH. Severe Heart Failure after Using Bortezomib in a Patient with Multiple Myeloma and Cardiac Amyloidosis of Normal Wall Thickness. ACTA CARDIOLOGICA SINICA 2024; 40:454-457. [PMID: 39045372 PMCID: PMC11261364 DOI: 10.6515/acs.202407_40(4).20240410a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/10/2024] [Indexed: 07/25/2024]
Affiliation(s)
- Ting-Wei Kao
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
| | - Tai-Chung Huang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
| | - Che-Wei Liao
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
- Department of Medicine, National Taiwan University Cancer Center
| | - Chia-Tung Shun
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Hsuan Tsai
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
| | - Yen-Hung Lin
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
| |
Collapse
|
34
|
Grynblat J, Khouri C, Hlavaty A, Jaïs X, Savale L, Chaumais MC, Kularatne M, Jevnikar M, Boucly A, Antigny F, Perros F, Simonneau G, Sitbon O, Humbert M, Montani D. Characteristics and outcomes of patients developing pulmonary hypertension associated with proteasome inhibitors. Eur Respir J 2024; 63:2302158. [PMID: 38697649 DOI: 10.1183/13993003.02158-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/15/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) has been described in patients treated with proteasome inhibitors (PIs). Our objective was to evaluate the association between PIs and PAH. METHODS Characteristics of incident PAH cases previously treated with carfilzomib or bortezomib were analysed from the French pulmonary hypertension registry and the VIGIAPATH programme from 2004 to 2023, concurrently with a pharmacovigilance disproportionality analysis using the World Health Organization (WHO) global database (VigiBase) and a meta-analysis of randomised controlled trials. RESULTS 11 incident cases of PI-associated PAH were identified (six with carfilzomib and five with bortezomib) with a female:male ratio of 2.7:1, a median age of 61 years, and a median delay between PI first exposure and PAH of 6 months. Four patients died (two from right heart failure, one from respiratory distress and one from an unknown cause). At diagnosis, six were in New York Heart Association Functional Class III/IV with severe haemodynamic impairment (median mean pulmonary arterial pressure 39 mmHg, cardiac index 2.45 L·min-1·m-2 and pulmonary vascular resistance 7.2 WU). In the WHO pharmacovigilance database, 169 cases of PH associated with PI were reported since 2013 with significant signals of disproportionate reporting (SDR) for carfilzomib, regardless of the definition of cases or control group. However, SDR for bortezomib were inconsistent. The systematic review identified 17 clinical trials, and carfilzomib was associated with a significantly higher risk of dyspnoea, severe dyspnoea and PH compared with bortezomib. CONCLUSION PIs may induce PAH in patients undergoing treatment, with carfilzomib emitting a stronger signal than bortezomib, and these patients should be monitored closely.
Collapse
Affiliation(s)
- Julien Grynblat
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
- These authors contributed equally to this work
| | - Charles Khouri
- Univ. Grenoble Alpes, HP2 Laboratory, INSERM U 1300, Grenoble, France
- Pharmacovigilance Unit and Clinical Pharmacology Department, Grenoble Alpes University Hospital, Grenoble, France
- These authors contributed equally to this work
| | - Alex Hlavaty
- Univ. Grenoble Alpes, HP2 Laboratory, INSERM U 1300, Grenoble, France
- Pharmacovigilance Unit and Clinical Pharmacology Department, Grenoble Alpes University Hospital, Grenoble, France
| | - Xavier Jaïs
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Laurent Savale
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Marie Camille Chaumais
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- School of Pharmacy, University of Paris-Saclay, Saclay, France
- AP-HP, Department of Pharmacy, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Mithum Kularatne
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
- Division of Respiratory Medicine, Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mitja Jevnikar
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Athénaïs Boucly
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Frédéric Perros
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Gérald Simonneau
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Olivier Sitbon
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - David Montani
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- School of Medicine, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
35
|
Costa BA, Costa TA, Saravia SD, Felix N, Tan CR, Korde N, Richter J. Thromboembolic risk of carfilzomib or bortezomib in combination with lenalidomide and dexamethasone for newly diagnosed multiple myeloma: A comparative systematic review and meta-analysis. Am J Hematol 2024; 99:1056-1065. [PMID: 38488702 DOI: 10.1002/ajh.27288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/06/2024] [Accepted: 03/04/2024] [Indexed: 05/16/2024]
Abstract
Thrombosis represents a frequent and potentially severe complication in individuals diagnosed with multiple myeloma (MM). These events can be driven by both the disease as well as the therapies themselves. Overall, available evidence is inconclusive about the differential thrombogenicity of carfilzomib/lenalidomide/dexamethasone (KRd) and bortezomib/lenalidomide/dexamethasone (VRd). This meta-analysis compares the risk for venous thromboembolism (VTE; including deep venous thrombosis and pulmonary embolism) and arterial thromboembolism (ATE; including myocardial infarction and ischemic stroke) with KRd versus VRd as primary therapy for newly diagnosed MM (NDMM). Out of 510 studies identified after deduplication, one randomized controlled trial and five retrospective cohort studies were included. We analyzed 2304 patients (VRd: 1380; KRd: 924) for VTE events and 2179 patients (VRd: 1316; KRd: 863) for ATE events. Lower rates of VTE were observed in the VRd group when compared with the KRd group (6.16% vs. 8.87%; odds ratio [OR], 0.53; 95% confidence interval [CI], 0.32-0.88; p = .01). Both treatment groups exhibited minimal ATE incidence, with no significant difference between them (0.91% vs. 1.16%; OR, 1.01; 95% CI, 0.24-4.20; p = .99). In view of potential biases from retrospective studies, heterogeneity of baseline population characteristics, and limited access to patient-level data (e.g., VTE risk stratification and type of thromboprophylaxis regimen used) inherent to this meta-analysis, additional research is warranted to further validate our findings and refine strategies for thrombosis prevention in MM.
Collapse
Affiliation(s)
- Bruno Almeida Costa
- Department of Medicine, Mount Sinai Morningside and West, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Medicine, Memorial Sloan Kettering Cancer, New York, New York, USA
| | | | - Sara Diaz Saravia
- Department of Medicine, Mount Sinai Morningside and West, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nicole Felix
- Department of Medicine, Federal University of Campina Grande, Campina Grande, Brazil
| | - Carlyn Rose Tan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer, New York, New York, USA
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer, New York, New York, USA
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Joshua Richter
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
36
|
Leiva O, Beaty W, Soo S, Agarwal MA, Yang EH. Cancer Therapy-Associated Pulmonary Hypertension and Right Ventricular Dysfunction: Etiologies and Prognostic Implications. Rev Cardiovasc Med 2024; 25:87. [PMID: 39076943 PMCID: PMC11263834 DOI: 10.31083/j.rcm2503087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 07/31/2024] Open
Abstract
Advances in cancer therapies have improved oncologic outcomes but can potentially expose patients to risk of cardiovascular toxicity. While left ventricular (LV) dysfunction is a well-known cardiotoxicity of cancer therapy. Pulmonary hypertension (PH) and right ventricular (RV) dysfunction are seen with several cancer therapies, including alkylating agents, tyrosine kinase inhibitors (TKIs), and immunotherapy, and are associated with significant morbidity and mortality. Awareness and recognition of cancer therapy-associated PH and RV dysfunction is critical to identify underlying etiologies and institute the appropriate therapy. However, gaps exist in the current literature on the epidemiology of PH and RV dysfunction in cancer, underlying pathophysiology and optimal management strategies.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - William Beaty
- Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Steven Soo
- Department of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Manyoo A. Agarwal
- Cardio-Oncology Program, Heart, Vascular and Thoracic Institute, Cleveland Clinic Abu Dhabi, 00000 Abu Dhabi, United Arab Emirates
| | - Eric H. Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
37
|
Pandey S, Tan EFS, Bellamkonda A, Aryal B, Kalavar M. Cyclophosphamide, Bortezomib, and Dexamethasone and Severe Systolic Heart Failure: A Case Report. Cureus 2024; 16:e56966. [PMID: 38665754 PMCID: PMC11044976 DOI: 10.7759/cureus.56966] [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: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Multiple myeloma (MM) is a neoplastic proliferation of plasma cells in bone marrow. Pharmacotherapy for the management of patients with MM includes drug classes like proteasome inhibitors, monoclonal antibodies, immunomodulators, alkylating agents, steroids, etc. We present a case of new-onset heart failure with reduced ejection fraction (HFrEF) in a patient with previously normal ejection fraction after treatment with a cyclophosphamide, bortezomib, and dexamethasone (CyBorD) chemotherapeutic regimen. An echocardiogram done after the completion of nine cycles of chemotherapy in a period of about 4.5 months showed severely decreased left ventricular systolic function with an ejection fraction of only 15-20% and grade I diastolic dysfunction. Cardiac catheterization showed no angiographic evidence of vessel occlusion or epicardial disease. HFrEF was managed with the initiation of guideline-directed medical therapy with cardiology clinic follow-up, and the patient was discharged with a plan to start a lenalidomide-based chemotherapeutic regimen with oncology clinic follow-up. It is, therefore, imperative to perform a thorough cardiovascular assessment before initiation of chemotherapy, complemented by periodic and recurrent assessments of cardiovascular function during and after completion of the treatment course, for early detection and prevention of potentially severe cardiovascular toxicities in patients with MM.
Collapse
Affiliation(s)
- Sagar Pandey
- Internal Medicine, One Brooklyn Health/Interfaith Medical Center, Brooklyn, USA
| | | | - Amulya Bellamkonda
- Internal Medicine, One Brooklyn Health/Interfaith Medical Center, Brooklyn, USA
| | - Binit Aryal
- Internal Medicine, One Brooklyn Health/Interfaith Medical Center, Brooklyn, USA
| | - Madhumati Kalavar
- Hematology and Oncology, One Brooklyn Health/Interfaith Medical Center, Brooklyn, USA
| |
Collapse
|
38
|
Jang B, Jeong J, Heo KN, Koh Y, Lee JY. Real-world incidence and risk factors of bortezomib-related cardiovascular adverse events in patients with multiple myeloma. Blood Res 2024; 59:3. [PMID: 38485811 PMCID: PMC10903519 DOI: 10.1007/s44313-024-00004-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Although most studies on the cardiovascular toxicity of proteasome inhibitors have focused on carfilzomib, the risk of cardiotoxicity associated with bortezomib remains controversial. This study aimed to evaluate the incidence and risk factors of cardiovascular adverse events (CVAEs) associated with bortezomib in patients with multiple myeloma in a real-world setting. METHODS This cross-sectional study included patients who were treated with bortezomib at a tertiary hospital in South Korea. CVAEs, defined as hypertension, arrhythmia, heart failure, myocardial infarction, pulmonary arterial hypertension, angina, and venous thromboembolism, were detected using cardiac markers, ECG, echocardiography, medications, or documentation by clinicians. The patients were observed for at least 6 months and up to 2 years after starting bortezomib administration. RESULTS Among the 395 patients, 20.8% experienced CVAEs of any grade, and 14.7% experienced severe adverse events. The median onset time for any CVAE was 101.5 days (IQR, 42-182 days), and new-onset/worsened hypertension was the most prevalent CVAE. The risk of CVAEs increased in patients with a body mass index lower than 18.5 (adjusted HR (aHR) 3.50, 95% confidence interval (CI) 1.05-11.72), light chain (1.80, 1.04-3.13), and IgD (4.63, 1.06-20.20) as the multiple myeloma subtype, baseline stroke (4.52, 1.59-12.80), and hypertension (1.99, 1.23-3.23). However, CVAEs did not significantly affect the 2-year overall survival and progression-free survival. CONCLUSION Approximately 15% of the Korean patients treated with bortezomib experienced severe CVAEs. Thus, patients, especially those with identified risk factors, should be closely monitored for CVAE symptoms during bortezomib treatment.
Collapse
Affiliation(s)
- Bitna Jang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
- Department of Pharmacy, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jonghyun Jeong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Kyu-Nam Heo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University Hospital, Daehak-Ro Jongno-Gu, 101, Seoul, Republic of Korea.
| | - Ju-Yeun Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
39
|
Larsson P, Pettersson D, Olsson M, Sarathchandra S, Abramsson A, Zetterberg H, Ittner E, Forssell-Aronsson E, Kovács A, Karlsson P, Helou K, Parris TZ. Repurposing proteasome inhibitors for improved treatment of triple-negative breast cancer. Cell Death Discov 2024; 10:57. [PMID: 38286854 PMCID: PMC10825133 DOI: 10.1038/s41420-024-01819-5] [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: 06/04/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/31/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is associated with poor prognosis and limited treatment options due to the lack of important receptors (estrogen receptor [ER], progesterone receptor [PR], and human epidermal growth factor receptor 2 [HER2]) used for targeted therapy. However, high-throughput in vitro drug screening of cell lines is a powerful tool for identifying effective drugs for a disease. Here, we determine the intrinsic chemosensitivity of TNBC cell lines to proteasome inhibitors (PIs), thereby identifying potentially potent 2-drug combinations for TNBC. Eight TNBC cell lines (BT-549, CAL-148, HCC1806, HCC38, HCC70, MDA-MB-436, MDA-MB-453, and MDA-MB-468) and two controls (MCF-10A and MCF-7) were first exposed to 18 drugs (11 PIs and 7 clinically relevant chemotherapeutic agents) as monotherapy, followed by prediction of potent 2-drug combinations using the IDACombo pipeline. The synergistic effects of the 2-drug combinations were evaluated with SynergyFinder in four TNBC cell lines (CAL-148, HCC1806, HCC38, and MDA-MB-468) and three controls (BT-474, MCF-7, and T47D) in vitro, followed by further evaluation of tumor regression in zebrafish tumor models established using HCC1806 and MCF-7 cells. Monotherapy identified nine effective drugs (bortezomib, carfilzomib, cisplatin, delanzomib, docetaxel, epoxomicin, MLN-2238, MLN-9708, and nedaplatin) across all cell lines. PIs (e.g., bortezomib, delanzomib, and epoxomicin) were highly potent drugs in TNBC cells, of which bortezomib and delanzomib inhibited the chymotrypsin-like activity of the 20 S proteasome by 100% at 10 µM. Moreover, several potent 2-drug combinations (e.g., bortezomib+nedaplatin and epoxomicin+epirubicin) that killed virtually 100% of cells were also identified. Although HCC1806- and MCF-7-derived xenografts treated with bortezomib+nedaplatin and carboplatin+paclitaxel were smaller, HCC1806 cells frequently metastasized to the trunk region. Taken together, we show that PIs used in combination with platinum agents or topoisomerase inhibitors exhibit increased efficiency with almost 100% inhibition in TNBC cell lines, indicating that PIs are therefore promising compounds to use as combination therapy for TNBC.
Collapse
Affiliation(s)
- Peter Larsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Daniella Pettersson
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maxim Olsson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | | | - Alexandra Abramsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Dementia Research Institute, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ella Ittner
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eva Forssell-Aronsson
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Khalil Helou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
40
|
Pandey S, Kalaria A, Jhaveri KD, Herrmann SM, Kim AS. Management of hypertension in patients with cancer: challenges and considerations. Clin Kidney J 2023; 16:2336-2348. [PMID: 38046043 PMCID: PMC10689173 DOI: 10.1093/ckj/sfad195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Indexed: 12/05/2023] Open
Abstract
The survival rates of many cancers have significantly improved due to recent advancements in cancer screening and therapeutics. Although better cancer outcomes are encouraging, additional health challenges have surfaced, the utmost of which is the burden imposed by various cardiovascular and renal toxicities of anticancer therapies. To improve the overall outcome of patients with cancer, it is essential to understand and manage these treatment-related adverse effects. The cardiovascular side effects of antineoplastic therapies are well-known and include left ventricular dysfunction, heart failure, myocardial ischaemia, QT prolongation, arrhythmia and hypertension. Among these, hypertension is the most common complication, prevalent in about 40% of all cancer patients, yet frequently overlooked and undertreated. This review explores the intricate connection between cancer and hypertension and provides distinct approaches to diagnosing, monitoring and managing hypertension in patients with cancer. We also outline the challenges and considerations that are relevant to the care of patients receiving anticancer drugs with prohypertensive potential.
Collapse
Affiliation(s)
- Shubhi Pandey
- Department of Internal Medicine, Calhoun Cardiology Center, University of Connecticut Health, Farmington, CT, USA
- University of Connecticut School of Medicine, Farmington, CT, USA
| | - Amar Kalaria
- University of Connecticut School of Medicine, Farmington, CT, USA
| | - Kenar D Jhaveri
- Division of Kidney Diseases and Hypertension, Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Agnes S Kim
- Department of Internal Medicine, Calhoun Cardiology Center, University of Connecticut Health, Farmington, CT, USA
- University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
41
|
Patiño-Escobar B, Talbot A, Wiita AP. Overcoming proteasome inhibitor resistance in the immunotherapy era. Trends Pharmacol Sci 2023; 44:507-518. [PMID: 37344251 DOI: 10.1016/j.tips.2023.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023]
Abstract
Proteasome inhibitors (PIs) are a fascinating class of small molecules that disrupt protein homeostasis and are highly efficacious in the blood cancer multiple myeloma. However, PIs are not curative, and overcoming PI resistance to extend patient survival remains a major unmet need. Recent strategies to overcome PI resistance, including inhibiting alternative protein homeostasis pathways and targeting the mitochondrion as a nexus of metabolic adaptation to PIs, are gaining momentum. However, these focused approaches may be surpassed or even obviated by quickly emerging immunotherapy strategies that do not selectively target PI resistance mechanisms but are highly efficacious in PI-resistant disease, nonetheless. Informed by insights from these promising areas of research moving in parallel, we propose that pharmacological strategies to enforce immunotherapeutic vulnerabilities in resistant disease may provide a unified outlook to overcome PI resistance in a 'new era' of myeloma treatment.
Collapse
Affiliation(s)
- Bonell Patiño-Escobar
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Alexis Talbot
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; INSERM U976, Institut de Recherche Saint Louis, Université de Paris, Paris, France
| | - Arun P Wiita
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA.
| |
Collapse
|
42
|
Lira JR, Guymon AL, Yang L, Sternburg JO, Giri S, Wang X. The double-hit protocol induces HFpEF and impairs myocardial ubiquitin-proteasome system performance in FVB/N mice. Front Physiol 2023; 14:1208153. [PMID: 37362441 PMCID: PMC10285383 DOI: 10.3389/fphys.2023.1208153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a leading cause of death and disability, with its prevalence surpassing that of heart failure with reduced ejection fraction. Obesity and hypertension are often associated with HFpEF. HFpEF can be modeled through simultaneous metabolic and hypertensive stresses in male C57BL/6N mice provoked by a combination treatment of a high-fat diet (HFD) and constitutive nitric oxide synthase inhibition by Nω-nitro-L-arginine methyl-ester (L-NAME). Ubiquitin-proteasome system (UPS) dysfunction was detected in many forms of cardiomyopathy, but whether it occurs in HFpEF remains unknown. We report successful modeling of HFpEF in male FVB/N mice and, by taking advantage of a transgenic UPS reporter mouse, we have detected myocardial UPS functioning impairment during HFpEF, suggesting a pathogenic role for impaired protein degradation in the development and progression of HFpEF.
Collapse
|