101
|
Touyz RM, Herrmann J. Cardiotoxicity with vascular endothelial growth factor inhibitor therapy. NPJ Precis Oncol 2018; 2:13. [PMID: 30202791 PMCID: PMC5988734 DOI: 10.1038/s41698-018-0056-z] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis inhibitors targeting the vascular endothelial growth factor (VEGF) signaling pathway (VSP) have been important additions in the therapy of various cancers, especially renal cell carcinoma and colorectal cancer. Bevazicumab, the first VSP to receive FDA approval in 2004 targeting all circulating isoforms of VEGF-A, has become one of the best-selling drugs of all times. The second wave of tyrosine kinase inhibitors (TKIs), which target the intracellular site of VEGF receptor kinases, began with the approval of sorafenib in 2005 and sunitinib in 2006. Heart failure was subsequently noted, in 2-4% of patients on bevacizumab and in 3-8% of patients on VSP-TKIs. The very fact that the single-targeted monoclonal antibody bevacizumab can induce cardiotoxicity supports a pathomechanistic role for the VSP and the postulate of the "vascular" nature of VSP inhibitor cardiotoxicity. In this review we will outline this scenario in greater detail, reflecting on hypertension and coronary artery disease as risk factors for VSP inhibitor cardiotoxicity, but also similarities with peripartum and diabetic cardiomyopathy. This leads to the concept that any preexisting or coexisting condition that reduces the vascular reserve or utilizes the vascular reserve for compensatory purposes may pose a risk factor for cardiotoxicity with VSP inhibitors. These conditions need to be carefully considered in cancer patients who are to undergo VSP inhibitor therapy. Such vigilance is not to exclude patients from such prognostically extremely important therapy but to understand the continuum and to recognize and react to any cardiotoxicity dynamics early on for superior overall outcomes.
Collapse
Affiliation(s)
- Rhian M. Touyz
- Institute of Cardiovascular & Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
102
|
|
103
|
Vrettos EI, Mező G, Tzakos AG. On the design principles of peptide-drug conjugates for targeted drug delivery to the malignant tumor site. Beilstein J Org Chem 2018; 14:930-954. [PMID: 29765474 PMCID: PMC5942387 DOI: 10.3762/bjoc.14.80] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/04/2018] [Indexed: 12/30/2022] Open
Abstract
Cancer is the second leading cause of death affecting nearly one in two people, and the appearance of new cases is projected to rise by >70% by 2030. To effectively combat the menace of cancer, a variety of strategies have been exploited. Among them, the development of peptide–drug conjugates (PDCs) is considered as an inextricable part of this armamentarium and is continuously explored as a viable approach to target malignant tumors. The general architecture of PDCs consists of three building blocks: the tumor-homing peptide, the cytotoxic agent and the biodegradable connecting linker. The aim of the current review is to provide a spherical perspective on the basic principles governing PDCs, as also the methodology to construct them. We aim to offer basic and integral knowledge on the rational design towards the construction of PDCs through analyzing each building block, as also to highlight the overall progress of this rapidly growing field. Therefore, we focus on several intriguing examples from the recent literature, including important PDCs that have progressed to phase III clinical trials. Last, we address possible difficulties that may emerge during the synthesis of PDCs, as also report ways to overcome them.
Collapse
Affiliation(s)
- Eirinaios I Vrettos
- University of Ioannina, Department of Chemistry, Section of Organic Chemistry and Biochemistry, Ioannina, GR-45110, Greece
| | - Gábor Mező
- Eötvös Loránd University, Faculty of Science, Institute of Chemistry, Pázmány P. stny. 1/A, H-1117 Budapest, Hungary.,MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, Pázmány P. stny. 1/A, H-1117 Budapest, Hungary
| | - Andreas G Tzakos
- University of Ioannina, Department of Chemistry, Section of Organic Chemistry and Biochemistry, Ioannina, GR-45110, Greece
| |
Collapse
|
104
|
Truitt R, Mu A, Corbin EA, Vite A, Brandimarto J, Ky B, Margulies KB. Increased Afterload Augments Sunitinib-Induced Cardiotoxicity in an Engineered Cardiac Microtissue Model. JACC Basic Transl Sci 2018; 3:265-276. [PMID: 30062212 PMCID: PMC6059907 DOI: 10.1016/j.jacbts.2017.12.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/22/2017] [Accepted: 12/22/2017] [Indexed: 12/17/2022]
Abstract
Sunitinib, a multitargeted oral tyrosine kinase inhibitor, used widely to treat solid tumors, results in hypertension in up to 47% and left ventricular dysfunction in up to 19% of treated individuals. The relative contribution of afterload toward inducing cardiac dysfunction with sunitinib treatment remains unknown. We created a preclinical model of sunitinib cardiotoxicity using engineered microtissues that exhibited cardiomyocyte death, decreases in force generation, and spontaneous beating at clinically relevant doses. Simulated increases in afterload augmented sunitinib cardiotoxicity in both rat and human microtissues, which suggest that antihypertensive therapy may be a strategy to prevent left ventricular dysfunction in patients treated with sunitinib.
Collapse
Key Words
- 2D, 2-dimensional
- 3D, 3-dimensional
- AICAR, 5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside
- AMPK, adenosine monophosphate-activated protein kinase
- ATP, adenosine triphosphate
- CCCP, carbonyl cyanide m-chlorophenyl hydrazine
- CMT, cardiac microtissue
- DMSO, dimethyl sulfoxide
- EDTA, ethylenediamine tetraacetic acid
- Hu-iPS-CM, human induced pluripotent stem cell cardiomyocyte
- LV, left ventricle
- NRVM, neonatal rat ventricular myocyte
- PDMS, polydimethylsiloxane
- RPMI, Roswell Park Memorial Institute medium
- TMRM, tetramethylrhodamine
- afterload
- apoptosis
- cardiotoxicity
- huMSC, human mesenchymal stem cell
- iPS-CM, induced pluripotent stem cell-derived cardiomyocyte
- sunitinib
- tissue engineering
- toxicology
- tyrosine kinase inhibitors
Collapse
Affiliation(s)
- Rachel Truitt
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anbin Mu
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elise A. Corbin
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Mechanical Engineering and Applied Mechanics, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexia Vite
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey Brandimarto
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bonnie Ky
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kenneth B. Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
105
|
Argyros O, Karampelas T, Varela A, Asvos X, Papakyriakou A, Agalou A, Beis D, Davos CH, Fokas D, Tamvakopoulos C. Targeting of the breast cancer microenvironment with a potent and linkable oxindole based antiangiogenic small molecule. Oncotarget 2018; 8:37250-37262. [PMID: 28422745 PMCID: PMC5514907 DOI: 10.18632/oncotarget.16763] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 03/22/2017] [Indexed: 01/11/2023] Open
Abstract
The clinical efficacy of antiangiogenic small molecules (e.g., sunitinib) in breast carcinoma has largely failed with substantial off-target toxicity. We rationally designed and evaluated preclinically a novel sunitinib analogue, SAP, with favourable pharmacological properties and the ability to be readily conjugated to a targeting peptide or antibody for active tumour targeting. SAP was evaluated in silico and in vitro in order to verify target engagement (e.g., VEGFR2). Pharmacokinetic and biodistribution parameters were determined in mice using LC-MS/MS. SAP efficacy was tested in two breast cancer xenograft and two syngeneic animal models and pharmacodynamic evaluation was accomplished using phosphokinase assays and immunohistochemistry. Cardiac and blood toxicity of SAP were also monitored. SAP retained the antiangiogenic and cytotoxic properties of the parental molecule with an increased blood exposure and tumor accumulation compared to sunitinib. SAP proved efficacious in all animal models. Tumors from SAP treated animals had significantly decreased Ki-67 and CD31 markers and reduced levels of phosphorylated AKT, ERK and S6 compared to vehicle treated animals. In mice dosed with SAP there was negligible hematotoxicity, while cardiac function measurements showed a reduction in the percentage left ventricular fractional shortening compared to vehicle treated animals. In conclusion, SAP is a novel rationally designed conjugatable small antiangiogenic molecule, efficacious in preclinical models of breast cancer.
Collapse
Affiliation(s)
- Orestis Argyros
- Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation Academy of Athens, Athens, 11527, Greece
| | - Theodoros Karampelas
- Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation Academy of Athens, Athens, 11527, Greece
| | - Aimilia Varela
- Cardiovascular Research Laboratory, Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, Athens, 11527, Greece
| | - Xenophon Asvos
- Laboratory of Medicinal Chemistry, Department of Materials Science and Engineering, University of Ioannina, Ioannina, 45110, Greece
| | - Athanasios Papakyriakou
- Laboratory of Chemical Biology of Natural Products and Designed Molecules, Institute of Physical Chemistry, N.C.S.R "Demokritos", Athens, 15310, Greece
| | - Adamantia Agalou
- Developmental Biology, Biomedical Research Foundation Academy of Athens, Athens, 11527, Greece
| | - Dimitris Beis
- Developmental Biology, Biomedical Research Foundation Academy of Athens, Athens, 11527, Greece
| | - Constantinos H Davos
- Cardiovascular Research Laboratory, Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, Athens, 11527, Greece
| | - Demosthenes Fokas
- Laboratory of Medicinal Chemistry, Department of Materials Science and Engineering, University of Ioannina, Ioannina, 45110, Greece
| | - Constantin Tamvakopoulos
- Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation Academy of Athens, Athens, 11527, Greece
| |
Collapse
|
106
|
Touyz RM, Herrmann SMS, Herrmann J. Vascular toxicities with VEGF inhibitor therapies-focus on hypertension and arterial thrombotic events. ACTA ACUST UNITED AC 2018; 12:409-425. [PMID: 29703600 PMCID: PMC6168784 DOI: 10.1016/j.jash.2018.03.008] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/02/2018] [Accepted: 03/14/2018] [Indexed: 12/21/2022]
Abstract
The vascular endothelial growth factor (VEGF) signaling pathway (VSP) fulfills a cardinal role in endothelial cells and its inhibition has profound cardiovascular impact. This is true not only for the normal vasculature but also for the tumor vasculature when VSP inhibitors are used as anti-angiogenic therapies. Generalized endothelial dysfunction predisposes to vasoconstriction, atherosclerosis, platelet activation, and thrombosis (arterial more than venous). All of these have been reported with VSP inhibitors and collectively give rise to vascular toxicities, the most concerning of which are arterial thromboembolic events (ATE). VSP inhibitors include antibodies, acting extracelluarly on VEGF, such as bevacizumab and tyrosine kinases inhibitors, acting intracellularly on the kinase domain of VEGF receptors, such as sunintib and sorafenib. The addition of bevacizumab and VSP tyrosine kinase inhibitor therapy to the cancer treatment regimen is associated with a 1.5-2.5-fold and 2.3-4.6-fold increase risk of ATEs, respectively. Risk factors for ATEs while on VSP inhibitor therapy include age older than 65 years, previous thromboembolic events, history of atherosclerotic disease, and duration of VSP inhibitor therapy. In clinical practice, hypertension remains the most commonly noted vascular manifestation of VSP inhibition. Optimal blood pressure goals and preferred therapeutic strategies toward reaching these goals are not defined at present. This review summarizes current data on this topic and proposes a more intensive management approach to patients undergoing VSP inhibitor therapy including Systolic Blood PRessure Intervention Trial (SPRINT) blood pressure goals, pleiotropic vasoprotective agents such as angiotensin converting enzyme inhibitors, amlodipine, and carvedilol, high-dose statin therapy, and aspirin.
Collapse
Affiliation(s)
- Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Sandra M S Herrmann
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
107
|
Chemotherapeutic-Induced Cardiovascular Dysfunction: Physiological Effects, Early Detection-The Role of Telomerase to Counteract Mitochondrial Defects and Oxidative Stress. Int J Mol Sci 2018. [PMID: 29534446 PMCID: PMC5877658 DOI: 10.3390/ijms19030797] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although chemotherapeutics can be highly effective at targeting malignancies, their ability to trigger cardiovascular morbidity is clinically significant. Chemotherapy can adversely affect cardiovascular physiology, resulting in the development of cardiomyopathy, heart failure and microvascular defects. Specifically, anthracyclines are known to cause an excessive buildup of free radical species and mitochondrial DNA damage (mtDNA) that can lead to oxidative stress-induced cardiovascular apoptosis. Therefore, oncologists and cardiologists maintain a network of communication when dealing with patients during treatment in order to treat and prevent chemotherapy-induced cardiovascular damage; however, there is a need to discover more accurate biomarkers and therapeutics to combat and predict the onset of cardiovascular side effects. Telomerase, originally discovered to promote cellular proliferation, has recently emerged as a potential mechanism to counteract mitochondrial defects and restore healthy mitochondrial vascular phenotypes. This review details mechanisms currently used to assess cardiovascular damage, such as C-reactive protein (CRP) and troponin levels, while also unearthing recently researched biomarkers, including circulating mtDNA, telomere length and telomerase activity. Further, we explore a potential role of telomerase in the mitigation of mitochondrial reactive oxygen species and maintenance of mtDNA integrity. Telomerase activity presents a promising indicator for the early detection and treatment of chemotherapy-derived cardiac damage.
Collapse
|
108
|
Varricchi G, Ameri P, Cadeddu C, Ghigo A, Madonna R, Marone G, Mercurio V, Monte I, Novo G, Parrella P, Pirozzi F, Pecoraro A, Spallarossa P, Zito C, Mercuro G, Pagliaro P, Tocchetti CG. Antineoplastic Drug-Induced Cardiotoxicity: A Redox Perspective. Front Physiol 2018; 9:167. [PMID: 29563880 PMCID: PMC5846016 DOI: 10.3389/fphys.2018.00167] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/20/2018] [Indexed: 12/28/2022] Open
Abstract
Antineoplastic drugs can be associated with several side effects, including cardiovascular toxicity (CTX). Biochemical studies have identified multiple mechanisms of CTX. Chemoterapeutic agents can alter redox homeostasis by increasing the production of reactive oxygen species (ROS) and reactive nitrogen species RNS. Cellular sources of ROS/RNS are cardiomyocytes, endothelial cells, stromal and inflammatory cells in the heart. Mitochondria, peroxisomes and other subcellular components are central hubs that control redox homeostasis. Mitochondria are central targets for antineoplastic drug-induced CTX. Understanding the mechanisms of CTX is fundamental for effective cardioprotection, without compromising the efficacy of anticancer treatments. Type 1 CTX is associated with irreversible cardiac cell injury and is typically caused by anthracyclines and conventional chemotherapeutic agents. Type 2 CTX, associated with reversible myocardial dysfunction, is generally caused by biologicals and targeted drugs. Although oxidative/nitrosative reactions play a central role in CTX caused by different antineoplastic drugs, additional mechanisms involving directly and indirectly cardiomyocytes and inflammatory cells play a role in cardiovascular toxicities. Identification of cardiologic risk factors and an integrated approach using molecular, imaging, and clinical data may allow the selection of patients at risk of developing chemotherapy-related CTX. Although the last decade has witnessed intense research related to the molecular and biochemical mechanisms of CTX of antineoplastic drugs, experimental and clinical studies are urgently needed to balance safety and efficacy of novel cancer therapies.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, Naples, Italy
| | - Pietro Ameri
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Christian Cadeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Rosalinda Madonna
- Institute of Cardiology, Center of Excellence on Aging, Università degli Studi “G. d'Annunzio” Chieti – Pescara, Chieti, Italy
- Department of Internal Medicine, Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, University of Texas Health Science Center, Houston, TX, United States
| | - Giancarlo Marone
- Section of Hygiene, Department of Public Health, University of Naples Federico II, Naples, Italy
- Monaldi Hospital Pharmacy, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Ines Monte
- Department of General Surgery and Medical-Surgery Specialities, University of Catania, Catania, Italy
| | - Giuseppina Novo
- U.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa, Italy
| | - Paolo Parrella
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Antonio Pecoraro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Paolo Spallarossa
- Clinic of Cardiovascular Diseases, IRCCS San Martino IST, Genova, Italy
| | - Concetta Zito
- Division of Clinical and Experimental Cardiology, Department of Medicine and Pharmacology, Policlinico “G. Martino” University of Messina, Messina, Italy
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
109
|
Yun J, Jeong H, Kim K, Han MH, Lee EH, Lee K, Cho C. β‐Adrenergic receptor agonists attenuate pericyte loss in diabetic retinas through Akt activation. FASEB J 2017; 32:2324-2338. [DOI: 10.1096/fj.201700570rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Jang‐Hyuk Yun
- Vascular Microenvironment Laboratory Department of Pharmacology Seoul National UniversitySeoul South Korea
| | - Han‐Seok Jeong
- Vascular Microenvironment Laboratory Department of Pharmacology Seoul National UniversitySeoul South Korea
| | - Kyung‐Jin Kim
- Vascular Microenvironment Laboratory Department of Pharmacology Seoul National UniversitySeoul South Korea
| | - Man Hyup Han
- Vascular Microenvironment Laboratory Department of Pharmacology Seoul National UniversitySeoul South Korea
| | - Eun Hui Lee
- Department of Physiology College of Medicine The Catholic University of Korea Seoul South Korea
| | - Kihwang Lee
- Department of Ophthalmology Ajou University School of Medicine Suwon South Korea
| | - Chung‐Hyun Cho
- Vascular Microenvironment Laboratory Department of Pharmacology Seoul National UniversitySeoul South Korea
| |
Collapse
|
110
|
Kempton A, Justice C, Guo A, Cefalu M, Makara M, Janssen P, Ho TH, Smith SA. Pazopanib for renal cell carcinoma leads to elevated mean arterial pressures in a murine model. Clin Exp Hypertens 2017; 40:524-533. [PMID: 29172746 DOI: 10.1080/10641963.2017.1403623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND In the setting of metastatic RCC (mRCC), pazopanib is approved as first line therapy. Unfortunately treatment may lead to cardiotoxicity such as hypertension, heart failure, and myocardial ischemia. OBJECTIVE Define the in vivo role of pazopanib in the development of cardiotoxicity. METHODS Wild type mice were dosed for 42 days via oral gavage, and separated into control and treatment (pazopanib) groups. Baseline ECG's, echocardiograms, and blood pressures were recorded. At the conclusion of the study functional parameters were again recorded, and animals were used for pathological, histological, and protein analysis. RESULTS After 2 weeks of dosing with pazopanib, the treatment group exhibited a statistically significant increase in mean arterial pressure compared to control mice (119 ± 11.7 mmHg versus 108 ± 8.2 mmHg, p = 0.049). Treatment with pazopanib led to a significant reduction in the cardiac output of mice. CONCLUSION Our findings in mice clearly demonstrate that treatment with pazopanib leads to a significant elevation in blood pressure after 2 weeks of dosing and this persists for the duration of dosing. The continued development of the cardio-oncology field will be paramount in providing optimal oncologic care while simultaneously improving cardiac outcomes through further investigation into the mechanisms of CV toxicity.
Collapse
Affiliation(s)
- Amber Kempton
- c Internal Medicine for Kempton, Davis Heart and Lung Research Institute, The Ohio State University , Columbus , OH , USA
| | - Cody Justice
- c Internal Medicine for Kempton, Davis Heart and Lung Research Institute, The Ohio State University , Columbus , OH , USA
| | - Aaron Guo
- c Internal Medicine for Kempton, Davis Heart and Lung Research Institute, The Ohio State University , Columbus , OH , USA
| | - Matthew Cefalu
- c Internal Medicine for Kempton, Davis Heart and Lung Research Institute, The Ohio State University , Columbus , OH , USA
| | - Michael Makara
- c Internal Medicine for Kempton, Davis Heart and Lung Research Institute, The Ohio State University , Columbus , OH , USA.,e Department of Medicine , Duke University School of Medicine , Durham , NC , USA
| | - Paul Janssen
- b Department of Physiology and Cell Biology , The Ohio State University , Columbus , OH , USA
| | - Thai H Ho
- d Internal Medicine (Oncology), Mayo Clinic Arizona , Phoenix , AZ , USA
| | - Sakima A Smith
- a Department of Internal Medicine (Division of Cardiology) , The Ohio State University College of Medicine , Columbus , OH , USA.,c Internal Medicine for Kempton, Davis Heart and Lung Research Institute, The Ohio State University , Columbus , OH , USA
| |
Collapse
|
111
|
Sethi TK, Basdag B, Bhatia N, Moslehi J, Reddy NM. Beyond Anthracyclines: Preemptive Management of Cardiovascular Toxicity in the Era of Targeted Agents for Hematologic Malignancies. Curr Hematol Malig Rep 2017; 12:257-267. [PMID: 28233150 DOI: 10.1007/s11899-017-0369-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Advances in drug discovery have led to the use of effective targeted agents in the treatment of hematologic malignancies. Drugs such as proteasome inhibitors in multiple myeloma and tyrosine kinase inhibitors in chronic myeloid leukemia and non-Hodgkin lymphoma have changed the face of treatment of hematologic malignancies. There are several reports of cardiovascular adverse events related to these newer agents. Both "on-target" and "off-target" effects of these agents can cause organ-specific toxicity. The need for long-term administration for most of these agents requires continued monitoring of toxicity. Moreover, the patient population is older, often over 50 years of age, making them more susceptible to cardiovascular side effects. Additional factors such as prior exposure to anthracyclines often add to this toxicity. In light of their success and widespread use, it is important to recognize and manage the unique side effect profile of targeted agents used in hematologic malignancies. In this article, we review the current data for the incidence of cardiovascular side effects of targeted agents in hematologic malignancies and discuss a preemptive approach towards managing these toxicities.
Collapse
Affiliation(s)
- Tarsheen K Sethi
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, 3927 The Vanderbilt Clinic, Nashville, TN, USA
| | - Basak Basdag
- Division of Internal Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nirmanmoh Bhatia
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Javid Moslehi
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nishitha M Reddy
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, 3927 The Vanderbilt Clinic, Nashville, TN, USA.
| |
Collapse
|
112
|
Junkunlo K, Söderhäll K, Noonin C, Söderhäll I. PDGF/VEGF-Related Receptor Affects Transglutaminase Activity to Control Cell Migration During Crustacean Hematopoiesis. Stem Cells Dev 2017; 26:1449-1459. [PMID: 28805145 DOI: 10.1089/scd.2017.0086] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The platelet-derived growth factor (PDGF) receptor, a tyrosine kinase (TK) receptor whose ligand is PDGF, is crucial in the transduction of extracellular signals into cells and mediates numerous processes, such as cell proliferation, differentiation, survival, and migration. We demonstrate the important roles of a receptor TK related to the PDGF/VEGF family protein (PVR) in controlling hematopoietic progenitor cell migration by affecting extracellular transglutaminase (TGase) activity. Pl_PVR1, GenBank accession No. KY444650, is highly expressed in hemocytes and the hematopoietic tissue (HPT). Sunitinib malate was used to block the PVF/PVR downstream pathway in HPT cell culture. The addition of Sunitinib also caused the HPT cells to increase in size and begin spreading. An increase in extracellular TGase activity on the HPT cell membrane was observed in a dose-dependent manner after treatment with Sunitinib malate. The presence of crude Ast1 provided a combinatorial beneficial effect that enhanced the number of spreading cells after inhibition of the Pl_PVR downstream signaling cascade. In addition, an increased immunoreactivity for β-tubulin and elongation of β-tubulin filaments were found in Pl_PVR signaling-inhibited cells. The potential roles of PVF/PVR signaling in controlling progenitor cell activity during hematopoiesis in crayfish were investigated and discussed.
Collapse
Affiliation(s)
- Kingkamon Junkunlo
- Department of Comparative Physiology, Uppsala University , Uppsala, Sweden
| | - Kenneth Söderhäll
- Department of Comparative Physiology, Uppsala University , Uppsala, Sweden
| | - Chadanat Noonin
- Department of Comparative Physiology, Uppsala University , Uppsala, Sweden
| | - Irene Söderhäll
- Department of Comparative Physiology, Uppsala University , Uppsala, Sweden
| |
Collapse
|
113
|
Jain D, Ahmad T, Cairo M, Aronow W. Cardiotoxicity of cancer chemotherapy: identification, prevention and treatment. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:348. [PMID: 28936442 PMCID: PMC5599271 DOI: 10.21037/atm.2017.06.35] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 06/07/2017] [Indexed: 12/21/2022]
Abstract
Cardiotoxicity is an important complication of several cancer therapeutic agents. Several well established and newer anticancer therapies such as anthracyclines, trastuzumab and other HER2 receptor blockers, antimetabolites, alkylating agents, tyrosine kinase inhibitors (TKIs), angiogenesis inhibitors, and checkpoint inhibitors are associated with significant cardiotoxicity. Cardiovascular imaging employing radionuclide imaging, echocardiography and magnetic resonance imaging are helpful in early detection and prevention of overt heart failure secondary to cardiotoxicity of cancer therapy. An understanding of the mechanism of the cardiotoxicity of cancer therapies can help prevent and treat their adverse cardiovascular consequences. Clinical implementation of algorithms based upon cardiac imaging and several non-imaging biomarkers can prevent cardiac morbidity and mortality associated with the use of cardiotoxic cancer therapies.
Collapse
Affiliation(s)
- Diwakar Jain
- Section of Cardiovascular Medicine, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Tauseef Ahmad
- Section of Oncology and Hematology, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Mitchel Cairo
- Section of Pediatric Hematology and Oncology, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Wilbert Aronow
- Section of Cardiovascular Medicine, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| |
Collapse
|
114
|
Abstract
The introduction of molecularly targeted therapies with tyrosine kinase inhibitors has revolutionized cancer therapy and has contributed to a steady decline in cancer-related mortality since the late 1990s. However, not only cardiac but also vascular toxicity has been reported for these agents, some as expected on-target effects (e.g., VEGF receptor inhibitors) and others as unanticipated events (e.g., BCR-Abl inhibitors). A sound understanding of these cardiovascular toxic effects is critical to advance mechanistic insight into vascular disease and clinical care. From a conceptual standpoint, there might be value in defining type I (permanent) and type II (transient) vascular toxicity. This review will focus on the tyrosine kinase inhibitors in current clinical use and their associated vascular side effects.
Collapse
|
115
|
Sourdon J, Lager F, Viel T, Balvay D, Moorhouse R, Bennana E, Renault G, Tharaux PL, Dhaun N, Tavitian B. Cardiac Metabolic Deregulation Induced by the Tyrosine Kinase Receptor Inhibitor Sunitinib is rescued by Endothelin Receptor Antagonism. Theranostics 2017; 7:2757-2774. [PMID: 28824714 PMCID: PMC5562214 DOI: 10.7150/thno.19551] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/31/2017] [Indexed: 02/06/2023] Open
Abstract
The growing field of cardio-oncology addresses the side effects of cancer treatment on the cardiovascular system. Here, we explored the cardiotoxicity of the antiangiogenic therapy, sunitinib, in the mouse heart from a diagnostic and therapeutic perspective. We showed that sunitinib induces an anaerobic switch of cellular metabolism within the myocardium which is associated with the development of myocardial fibrosis and reduced left ventricular ejection fraction as demonstrated by echocardiography. The capacity of positron emission tomography with [18F]fluorodeoxyglucose to detect the changes in cardiac metabolism caused by sunitinib was dependent on fasting status and duration of treatment. Pan proteomic analysis in the myocardium showed that sunitinib induced (i) an early metabolic switch with enhanced glycolysis and reduced oxidative phosphorylation, and (ii) a metabolic failure to use glucose as energy substrate, similar to the insulin resistance found in type 2 diabetes. Co-administration of the endothelin receptor antagonist, macitentan, to sunitinib-treated animals prevented both metabolic defects, restored glucose uptake and cardiac function, and prevented myocardial fibrosis. These results support the endothelin system in mediating the cardiotoxic effects of sunitinib and endothelin receptor antagonism as a potential therapeutic approach to prevent cardiotoxicity. Furthermore, metabolic and functional imaging can monitor the cardiotoxic effects and the benefits of endothelin antagonism in a theranostic approach.
Collapse
Affiliation(s)
- Joevin Sourdon
- Paris Cardiovascular Research Center (PARCC); INSERM UMR970; Université Paris Descartes; Paris, France
| | - Franck Lager
- Institut Cochin, Université Paris Descartes, INSERM U1016, Paris 75014, France
| | - Thomas Viel
- Paris Cardiovascular Research Center (PARCC); INSERM UMR970; Université Paris Descartes; Paris, France
| | - Daniel Balvay
- Paris Cardiovascular Research Center (PARCC); INSERM UMR970; Université Paris Descartes; Paris, France
| | - Rebecca Moorhouse
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Evangeline Bennana
- Institut Cochin, Université Paris Descartes, INSERM U1016, Paris 75014, France
- 3P5 proteomics facility, Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France
| | - Gilles Renault
- Institut Cochin, Université Paris Descartes, INSERM U1016, Paris 75014, France
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Research Center (PARCC); INSERM UMR970; Université Paris Descartes; Paris, France
| | - Neeraj Dhaun
- University/British Heart Foundation Centre of Research Excellence, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Bertrand Tavitian
- Paris Cardiovascular Research Center (PARCC); INSERM UMR970; Université Paris Descartes; Paris, France
- Service de Radiologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| |
Collapse
|
116
|
Harper A, Blackwood L. Toxicity and response in cats with neoplasia treated with toceranib phosphate. J Feline Med Surg 2017; 19:619-623. [PMID: 27090289 PMCID: PMC11128804 DOI: 10.1177/1098612x16643124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Objectives Toceranib phosphate is a tyrosine kinase inhibitor licensed for the treatment of non-resectable Patnaik grade II/III recurrent cutaneous mast cell tumours in dogs. There is no information in cats regarding the tolerated dose, toxicity or tumour response of this drug. The aim of this study was to analyse retrospectively a cohort of cats with advanced neoplasia treated with toceranib to identify toxicity and response. Methods The medical records of the Small Animal Teaching Hospital were reviewed. Cats were included if they had received toceranib for at least 2 weeks for the treatment of histologically or cytologically confirmed neoplastic disease, and had at least one set of monitoring blood tests (haematology, biochemistry) performed after baseline tests. Toxicity was graded according to the Veterinary Comparative Oncology Group - common terminology criteria for adverse events(VCOG-CTCAE) and response was measured according to Response Evaluation In Solid Tumors (RECIST) criteria. Results Fourteen cats met the inclusion criteria, the majority of which (13/14) had received previous therapy (surgery, radiotherapy, chemotherapy). The most common tumour types were mast cell tumours or malignant epithelial tumours. Toxicity occurred in 10/14 cats - 10 cats had mild myelosuppression or gastrointestinal effects. Two cats developed severe hepatoxicity. One cat died from congestive heart failure, although whether this was related to toceranib therapy is unknown. Regarding response, one cat achieved complete response; two cats achieved partial response and five cats achieved stable disease: overall biological response rate was 57.1%. All of the cats that achieved either partial or complete response were treated for mast cell disease. Overall median duration of response was 90 days (range 14-570 days). None of the cats with squamous cell carcinoma achieved a response. Conclusions and relevance Toceranib phosphate is generally well tolerated in cats, with toxicity limited to mild gastrointestinal or myelosuppressive effects in the majority of cases (10/14) in this study; however, hepatotoxicity is a concern. Response to treatment in this small cohort was similar to that reported in dogs.
Collapse
Affiliation(s)
- Aaron Harper
- Department of Veterinary Medicine, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
117
|
Tousoulis D. Cardio-oncology: Novel concepts and directions. Hellenic J Cardiol 2017; 58:175-177. [PMID: 28918283 DOI: 10.1016/j.hjc.2017.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 12/01/2022] Open
Affiliation(s)
- Dimitris Tousoulis
- 1st Cardiology Department, National and Kapodistrian University of Athens University Medical School, Hippokration Hospital, Athens, Greece.
| |
Collapse
|
118
|
Jain D, Russell RR, Schwartz RG, Panjrath GS, Aronow W. Cardiac Complications of Cancer Therapy: Pathophysiology, Identification, Prevention, Treatment, and Future Directions. Curr Cardiol Rep 2017; 19:36. [PMID: 28374177 DOI: 10.1007/s11886-017-0846-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Cardiotoxicity is an important complication of cancer therapy. With a significant improvement in the overall survival and prognosis of patients undergoing cancer therapy, cardiovascular toxicity of cancer therapy has become an important public health issue. Several well-established as well as newer anticancer therapies such as anthracyclines, trastuzumab, and other HER2 receptor blockers, antimetabolites, alkylating agents, tyrosine kinase inhibitors, angiogenesis inhibitors, checkpoint inhibitors, and thoracic irradiation are associated with significant cardiotoxicity. RECENT FINDINGS Cardiovascular imaging employing radionuclide imaging, echocardiography, and magnetic resonance imaging is helpful in early detection of the cardiotoxicity and prevention of overt heart failure. These techniques also provide important tools for understanding the mechanism of cardiotoxicity of these modalities, which would help develop strategies for the prevention of cardiac morbidity and mortality related to the use of these agents. An understanding of the mechanism of the cardiotoxicity of cancer therapies can help prevent and treat their adverse cardiovascular consequences. Clinical implementation of algorithms based upon cardiac imaging and several non-imaging biomarkers can prevent cardiac morbidity and mortality associated with the use of cardiotoxic cancer therapies.
Collapse
Affiliation(s)
- Diwakar Jain
- Section of Cardiovascular Medicine, New York Medical College and Westchester Medical Center, 100 Woods Road, Valhalla, NY, USA.
| | - Raymond R Russell
- Rhode Island Cardiovascular Institute, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Ronald G Schwartz
- Cardiology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Nuclear Medicine Division, Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - Gurusher S Panjrath
- Heart and Vascular Institute, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Wilbert Aronow
- Section of Cardiovascular Medicine, New York Medical College and Westchester Medical Center, 100 Woods Road, Valhalla, NY, USA
| |
Collapse
|
119
|
Tyrosine kinase-targeting drugs-associated heart failure. Br J Cancer 2017; 116:1366-1373. [PMID: 28399109 PMCID: PMC5482733 DOI: 10.1038/bjc.2017.88] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/17/2017] [Accepted: 02/23/2017] [Indexed: 02/02/2023] Open
Abstract
Background: The impact of cancer therapies on cardiac disease in the general adult cancer survivor population is largely unknown. Our objective was to evaluate which tyrosine kinase-targeting drugs are associated with greater risk for new-onset heart failure (HF). Methods: A nested case–control analysis was conducted within a cohort of 27 992 patients of Clalit Health Services, newly treated with a tyrosine kinase-targeting, and/or chemotherapeutic drug, for a malignant disease, between 1 January 2005 and 31 December 2012. Each new case of HF was matched to up to 30 controls from the cohort on calendar year of cohort entry, age, gender, and duration of follow-up. Main outcome measure was odds ratio (OR) with 95% confidence interval (CI) of new-onset HF. Results: There were 936 incident cases of HF during 71 742 person-years of follow-up. Trastuzumab (OR 1.90, 95% CI 1.46–2.49), cetuximab (OR 1.72, 1.10–2.69), panitumumab (OR 3.01, 1.02–8.85), and sunitinib (OR 3.39, 1.78–6.47) were associated with increased HF risk. Comorbidity independently associated with higher risk in a multivariable conditional regression model was diabetes mellitus, hypertension, chronic renal failure, ischaemic heart disease, valvular heart disease, arrhythmia, and smoking. Conclusions: Trastuzumab, cetuximab, panitumumab, and sunitinib are associated with increased risk for new-onset HF.
Collapse
|
120
|
O’Farrell AC, Evans R, Silvola JMU, Miller IS, Conroy E, Hector S, Cary M, Murray DW, Jarzabek MA, Maratha A, Alamanou M, Udupi GM, Shiels L, Pallaud C, Saraste A, Liljenbäck H, Jauhiainen M, Oikonen V, Ducret A, Cutler P, McAuliffe FM, Rousseau JA, Lecomte R, Gascon S, Arany Z, Ky B, Force T, Knuuti J, Gallagher WM, Roivainen A, Byrne AT. A Novel Positron Emission Tomography (PET) Approach to Monitor Cardiac Metabolic Pathway Remodeling in Response to Sunitinib Malate. PLoS One 2017; 12:e0169964. [PMID: 28129334 PMCID: PMC5271313 DOI: 10.1371/journal.pone.0169964] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 12/25/2016] [Indexed: 01/17/2023] Open
Abstract
Sunitinib is a tyrosine kinase inhibitor approved for the treatment of multiple solid tumors. However, cardiotoxicity is of increasing concern, with a need to develop rational mechanism driven approaches for the early detection of cardiac dysfunction. We sought to interrogate changes in cardiac energy substrate usage during sunitinib treatment, hypothesising that these changes could represent a strategy for the early detection of cardiotoxicity. Balb/CJ mice or Sprague-Dawley rats were treated orally for 4 weeks with 40 or 20 mg/kg/day sunitinib. Cardiac positron emission tomography (PET) was implemented to investigate alterations in myocardial glucose and oxidative metabolism. Following treatment, blood pressure increased, and left ventricular ejection fraction decreased. Cardiac [18F]-fluorodeoxyglucose (FDG)-PET revealed increased glucose uptake after 48 hours. [11C]Acetate-PET showed decreased myocardial perfusion following treatment. Electron microscopy revealed significant lipid accumulation in the myocardium. Proteomic analyses indicated that oxidative metabolism, fatty acid β-oxidation and mitochondrial dysfunction were among the top myocardial signalling pathways perturbed. Sunitinib treatment results in an increased reliance on glycolysis, increased myocardial lipid deposition and perturbed mitochondrial function, indicative of a fundamental energy crisis resulting in compromised myocardial energy metabolism and function. Our findings suggest that a cardiac PET strategy may represent a rational approach to non-invasively monitor metabolic pathway remodeling following sunitinib treatment.
Collapse
Affiliation(s)
- Alice C. O’Farrell
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Rhys Evans
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Johanna M. U. Silvola
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
| | - Ian S. Miller
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Emer Conroy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Suzanne Hector
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Roche Innovation Center Basel, F Hoffman La Roche, Basel, Switzerland
| | | | - David W. Murray
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Oncomark Ltd, Dublin, Ireland
| | - Monika A. Jarzabek
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Roche Innovation Center Basel, F Hoffman La Roche, Basel, Switzerland
| | | | | | | | - Liam Shiels
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Celine Pallaud
- Roche Innovation Center Basel, F Hoffman La Roche, Basel, Switzerland
| | - Antti Saraste
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
- Heart Center, Turku University Hospital and Åbo Akademi University, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
| | - Matti Jauhiainen
- Public Health Genomics Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Vesa Oikonen
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
| | - Axel Ducret
- Roche Innovation Center Basel, F Hoffman La Roche, Basel, Switzerland
| | - Paul Cutler
- Roche Innovation Center Basel, F Hoffman La Roche, Basel, Switzerland
| | - Fionnuala M. McAuliffe
- UCD Obstetrics & Gynaecology, School of Medicine, University College, Dublin, National Maternity Hospital, Dublin, Ireland
| | | | | | | | - Zoltan Arany
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, United States of America
| | - Bonnie Ky
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, United States of America
| | - Thomas Force
- Vanderbilt University School of Medicine, Nashville, United States of America
| | - Juhani Knuuti
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
| | - William M. Gallagher
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
- Oncomark Ltd, Dublin, Ireland
| | - Anne Roivainen
- Turku PET Centre, Turku University Hospital and Åbo Akademi University, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Annette T. Byrne
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- * E-mail:
| |
Collapse
|
121
|
Brown SA, Nhola L, Herrmann J. Cardiovascular Toxicities of Small Molecule Tyrosine Kinase Inhibitors: An Opportunity for Systems-Based Approaches. Clin Pharmacol Ther 2016; 101:65-80. [DOI: 10.1002/cpt.552] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 10/31/2016] [Accepted: 10/31/2016] [Indexed: 12/12/2022]
Affiliation(s)
- S-A Brown
- Department of Cardiovascular Diseases; Mayo Clinic; Rochester Minnesota USA
| | - L Nhola
- Department of Cardiovascular Diseases; Mayo Clinic; Rochester Minnesota USA
| | - J Herrmann
- Department of Cardiovascular Diseases; Mayo Clinic; Rochester Minnesota USA
| |
Collapse
|
122
|
Di Lisi D, Madonna R, Zito C, Bronte E, Badalamenti G, Parrella P, Monte I, Tocchetti CG, Russo A, Novo G. Anticancer therapy-induced vascular toxicity: VEGF inhibition and beyond. Int J Cardiol 2016; 227:11-17. [PMID: 27866063 DOI: 10.1016/j.ijcard.2016.11.174] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 11/06/2016] [Indexed: 11/27/2022]
Abstract
Cardiotoxicity induced by chemotherapeutic agents and radiotherapy is a growing problem. In recent years, an increasing number of new drugs with targeted action have been designed. These molecules, such as monoclonal antibodies and tyrosine kinase inhibitors, can cause different type of toxicities compared to traditional chemotherapy. However, they can also cause cardiac complications such as heart failure, arterial hypertension, QT interval prolongation and arrhythmias. Currently, a field of intense research is the vascular toxicity induced by new biologic drugs, particularly those which inhibit vascular endothelial growth factor (VEGF) and its receptor (VEGF-R) and other tyrosine kinases. In this review, we aim at focusing on the problem of vascular toxicity induced by new targeted therapies, chemotherapy and radiotherapy, and describe the main mechanisms and emphasizing the importance of early diagnosis of vascular damage, in order to prevent clinical complications.
Collapse
Affiliation(s)
- Daniela Di Lisi
- Division of Cardiology, Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy
| | - Rosalinda Madonna
- Center of Excellence on Aging, Institute of Cardiology, "G. d'Annunzio" University - Chieti, Chieti, Italy; Texas Heart Institute and University of Texas Medical School in Houston, Cardiology Division, Houston, TX, USA.
| | - Concetta Zito
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Enrico Bronte
- Department of Surgical, Oncological and Stomatological Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Stomatological Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Paolo Parrella
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, Naples, Italy
| | - Ines Monte
- Department of General Surgery and Medical-Surgery Specialties, University of Catania, Catania, Italy
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, Naples, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Stomatological Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppina Novo
- Division of Cardiology, Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy
| |
Collapse
|
123
|
Warmke N, Griffin KJ, Cubbon RM. Pericytes in diabetes-associated vascular disease. J Diabetes Complications 2016; 30:1643-1650. [PMID: 27592245 DOI: 10.1016/j.jdiacomp.2016.08.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/01/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022]
Abstract
Pericytes are mural cells that support and stabilise the microvasculature, and are present in all vascular beds. Pericyte-endothelial cell crosstalk is essential in both remodelling and quiescent vasculature, and this complex interaction is often disrupted in disease states. Pericyte loss is believed to be an early hallmark of diabetes-associated microvascular disease, including retinopathy and nephropathy. Here we review the current literature defining pericyte biology in the context of diabetes-associated vascular disease, with a particular focus on whether pericytes contribute actively to disease progression. We also speculate regarding the role of pericytes in the recovery from macrovascular complications, such as critical limb ischaemia. It becomes clear that dysfunctional pericytes are likely to actively induce disease progression by causing vasoconstriction and basement membrane thickening, resulting in tissue ischaemia. Moreover, their altered interactions with endothelial cells are likely to cause abnormal and inadequate neovascularisation in diverse vascular beds. Further research is needed to identify mechanisms by which pericyte function is altered by diabetes, with a view to developing therapeutic approaches that normalise vascular function and remodelling.
Collapse
Affiliation(s)
- Nele Warmke
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom.
| |
Collapse
|
124
|
Wilkinson EL, Sidaway JE, Cross MJ. Cardiotoxic drugs Herceptin and doxorubicin inhibit cardiac microvascular endothelial cell barrier formation resulting in increased drug permeability. Biol Open 2016; 5:1362-1370. [PMID: 27543060 PMCID: PMC5087671 DOI: 10.1242/bio.020362] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cardiotoxicity induced by anti-cancer therapeutics is a severe, and potentially fatal, adverse reaction of the heart in response to certain drugs. Current in vitro approaches to assess cardiotoxicity have focused on analysing cardiomyocytes. More recently it has become apparent that non-cardiomyocyte cells of the heart can potentially contribute to cardiotoxicity. Herceptin and doxorubicin are known to induce cardiotoxicity in the clinic. The effect of these drugs on the endothelial tight junction barrier was tested by analysing tight junction formation and zona occludens-1 (ZO-1) levels, revealing that Herceptin and doxorubicin are able to induce barrier perturbment and decrease barrier function in human cardiac microvascular endothelial cells (HCMECs) leading to increased permeability. Herceptin treatment had no effect on the tight junction barrier function in human dermal and human brain microvascular endothelial cells. HCMECs showed detectable levels of HER2 compared with the other endothelial cells suggesting that Herceptin binding to HER2 in these cells may interfere with tight junction formation. Our data suggests that doxorubicin and Herceptin can affect tight junction formation in the cardiac microvasculature leading to increased drug permeability and adverse effects on the cardiac myocytes. Summary: The anti-cancer drugs doxorubicin and Herceptin can disrupt tight junction formation in cardiac microvascular endothelial cells resulting in increased permeability which could potentially contribute to drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Emma L Wilkinson
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Building, Ashton Street, The University of Liverpool, Liverpool, L69 3GE, UK
| | - James E Sidaway
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Building, Ashton Street, The University of Liverpool, Liverpool, L69 3GE, UK Molecular Toxicology, AstraZeneca, Alderley Park, Cheshire, SK10 4TG, UK
| | - Michael J Cross
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Building, Ashton Street, The University of Liverpool, Liverpool, L69 3GE, UK
| |
Collapse
|
125
|
Caporali A, Martello A, Miscianinov V, Maselli D, Vono R, Spinetti G. Contribution of pericyte paracrine regulation of the endothelium to angiogenesis. Pharmacol Ther 2016; 171:56-64. [PMID: 27742570 DOI: 10.1016/j.pharmthera.2016.10.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
During physiological development and after a stressor event, vascular cells communicate with each other to evoke new vessel formation-a process known as angiogenesis. This communication occurs via direct contact and via paracrine release of proteins and nucleic acids, both in a free form or encapsulated into micro-vesicles. In diseases with an altered angiogenic response, such as cancer and diabetic vascular complications, it becomes of paramount importance to tune the cell communication process. Endothelial cell growth and migration are essential processes for new vessel formation, and pericytes, together with some classes of circulating monocytes, are important endothelial regulators. The interaction between pericytes and the endothelium is facilitated by their anatomical apposition, which involves endothelial cells and pericytes sharing the same basement membrane. However, the role of pericytes is not fully understood. The characteristics and the function of tissue-specific pericytesis are the focus of this review. Factors involved in the cross-talk between these cell types and the opportunities afforded by micro-RNA and micro-vesicle techniques are discussed. Targeting these mechanisms in pathological conditions, in which the vessel response is altered, is considered in relation to identification of new therapies for restoring the blood flow.
Collapse
Affiliation(s)
- A Caporali
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - A Martello
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - V Miscianinov
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - D Maselli
- IRCCS MultiMedica, Milan, Italy; Dipartimento di Scienze Biomediche, Università di Sassari, Sassari, Italy
| | - R Vono
- IRCCS MultiMedica, Milan, Italy
| | | |
Collapse
|
126
|
Michaloski JS, Redondo AR, Magalhães LS, Cambui CC, Giordano RJ. Discovery of pan-VEGF inhibitory peptides directed to the extracellular ligand-binding domains of the VEGF receptors. SCIENCE ADVANCES 2016; 2:e1600611. [PMID: 27819042 PMCID: PMC5091360 DOI: 10.1126/sciadv.1600611] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 09/27/2016] [Indexed: 05/03/2023]
Abstract
Receptor tyrosine kinases (RTKs) are key molecules in numerous cellular processes, the inhibitors of which play an important role in the clinic. Among them are the vascular endothelial growth factor (VEGF) family members and their receptors (VEGFR), which are essential in the formation of new blood vessels by angiogenesis. Anti-VEGF therapy has already shown promising results in oncology and ophthalmology, but one of the challenges in the field is the design of specific small-molecule inhibitors for these receptors. We show the identification and characterization of small 6-mer peptides that target the extracellular ligand-binding domain of all three VEGF receptors. These peptides specifically prevent the binding of VEGF family members to all three receptors and downstream signaling but do not affect other angiogenic RTKs and their ligands. One of the selected peptides was also very effective at preventing pathological angiogenesis in a mouse model of retinopathy, normalizing the vasculature to levels similar to those of a normal developing retina. Collectively, our results suggest that these peptides are pan-VEGF inhibitors directed at a common binding pocket shared by all three VEGFRs. These peptides and the druggable binding site they target might be important for the development of novel and selective small-molecule, extracellular ligand-binding inhibitors of RTKs (eTKIs) for angiogenic-dependent diseases.
Collapse
|
127
|
Croteau E, Renaud JM, Richard MA, Ruddy TD, Bénard F, deKemp RA. PET Metabolic Biomarkers for Cancer. BIOMARKERS IN CANCER 2016; 8:61-9. [PMID: 27679534 PMCID: PMC5030827 DOI: 10.4137/bic.s27483] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/08/2016] [Accepted: 05/19/2016] [Indexed: 02/06/2023]
Abstract
The body's main fuel sources are fats, carbohydrates (glucose), proteins, and ketone bodies. It is well known that an important hallmark of cancer cells is the overconsumption of glucose. Positron emission tomography (PET) imaging using the glucose analog (18)F-fluorodeoxyglucose ((18)F-FDG) has been a powerful cancer diagnostic tool for many decades. Apart from surgery, chemotherapy and radiotherapy represent the two main domains for cancer therapy, targeting tumor proliferation, cell division, and DNA replication-all processes that require a large amount of energy. Currently, in vivo clinical imaging of metabolism is performed almost exclusively using PET radiotracers that assess oxygen consumption and mechanisms of energy substrate consumption. This paper reviews the utility of PET imaging biomarkers for the detection of cancer proliferation, vascularization, metabolism, treatment response, and follow-up after radiation therapy, chemotherapy, and chemotherapy-related side effects.
Collapse
Affiliation(s)
- Etienne Croteau
- National Cardiac PET Centre, Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada; Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jennifer M Renaud
- National Cardiac PET Centre, Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Marie Anne Richard
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Terrence D Ruddy
- National Cardiac PET Centre, Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - François Bénard
- Division of Nuclear Medicine, Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Robert A deKemp
- National Cardiac PET Centre, Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
| |
Collapse
|
128
|
Murray IR, Baily JE, Chen WCW, Dar A, Gonzalez ZN, Jensen AR, Petrigliano FA, Deb A, Henderson NC. Skeletal and cardiac muscle pericytes: Functions and therapeutic potential. Pharmacol Ther 2016; 171:65-74. [PMID: 27595928 DOI: 10.1016/j.pharmthera.2016.09.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pericytes are periendothelial mesenchymal cells residing within the microvasculature. Skeletal muscle and cardiac pericytes are now recognized to fulfill an increasing number of functions in normal tissue homeostasis, including contributing to microvascular function by maintaining vessel stability and regulating capillary flow. In the setting of muscle injury, pericytes contribute to a regenerative microenvironment through release of trophic factors and by modulating local immune responses. In skeletal muscle, pericytes also directly enhance tissue healing by differentiating into myofibers. Conversely, pericytes have also been implicated in the development of disease states, including fibrosis, heterotopic ossication and calcification, atherosclerosis, and tumor angiogenesis. Despite increased recognition of pericyte heterogeneity, it is not yet clear whether specific subsets of pericytes are responsible for individual functions in skeletal and cardiac muscle homeostasis and disease.
Collapse
Affiliation(s)
- Iain R Murray
- BHF Center for Vascular Regeneration and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK; Department of Trauma and Orthopaedic Surgery, The University of Edinburgh, Edinburgh, UK
| | - James E Baily
- BHF Center for Vascular Regeneration and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - William C W Chen
- Reseach Laboratory of Electronics and Department of Biological Engineering, Massachusetts Institute of Technology, Boston, MA, USA
| | - Ayelet Dar
- Orthopedic Hospital Research Center, University of California, Los Angeles, CA, USA
| | - Zaniah N Gonzalez
- BHF Center for Vascular Regeneration and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Andrew R Jensen
- Orthopedic Hospital Research Center, University of California, Los Angeles, CA, USA
| | - Frank A Petrigliano
- Orthopedic Hospital Research Center, University of California, Los Angeles, CA, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine & Molecular Cell and Developmental Biology, and Eli and Edythe Broad Institute of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, CA, USA.
| | - Neil C Henderson
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
129
|
Herrmann J, Yang EH, Iliescu CA, Cilingiroglu M, Charitakis K, Hakeem A, Toutouzas K, Leesar MA, Grines CL, Marmagkiolis K. Vascular Toxicities of Cancer Therapies: The Old and the New--An Evolving Avenue. Circulation 2016; 133:1272-89. [PMID: 27022039 DOI: 10.1161/circulationaha.115.018347] [Citation(s) in RCA: 223] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Since the late 1990s, there has been a steady decline in cancer-related mortality, in part related to the introduction of so-called targeted therapies. Intended to interfere with a specific molecular pathway, these therapies have, paradoxically, led to a number of effects off their intended cancer tissue or molecular targets. The latest examples are tyrosine kinase inhibitors targeting the Philadelphia Chromosome mutation product, which have been associated with progressive atherosclerosis and acute vascular events. In addition, agents designed to interfere with the vascular growth factor signaling pathway have vascular side effects ranging from hypertension to arterial events and cardiomyocyte toxicity. Interestingly, the risk of cardiotoxicity with drugs such as trastuzumab is predicted by preexisting cardiovascular risk factors and disease, posing the question of a vascular component to the pathophysiology. The effect on the coronary circulation has been the leading explanation for the cardiotoxicity of 5-fluorouracil and may be the underlying the mechanism of presentation of apical ballooning syndrome with various chemotherapeutic agents. Classical chemotherapeutic agents such as cisplatin, often used in combination with bleomycin and vinca alkaloids, can lead to vascular events including acute coronary thrombosis and may be associated with an increased long-term cardiovascular risk. This review is intended to provide an update on the evolving spectrum of vascular toxicities with cancer therapeutics, particularly as they pertain to clinical practice, and to the conceptualization of cardiovascular diseases, as well. Vascular toxicity with cancer therapy: the old and the new, an evolving avenue.
Collapse
Affiliation(s)
- Joerg Herrmann
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.).
| | - Eric H Yang
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Cezar A Iliescu
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Mehmet Cilingiroglu
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Konstantinos Charitakis
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Abdul Hakeem
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Konstantinos Toutouzas
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Massoud A Leesar
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Cindy L Grines
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| | - Konstantinos Marmagkiolis
- From Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN (J.H.); University of California at Los Angeles, Division of Cardiology, Los Angeles (E.-H.Y.); University of Texas, MD Anderson Cancer Center, Houston (C.A.I.); Arkansas Heart Hospital, Little Rock, AR and Koc University School of Medicine, Istanbul, Turkey (M.C.); University of Texas Health Science Center, Houston (K.C.); University of Arkansas for Medical Sciences, Little Rock (A.H.); Athens Medical School, Hippokration General Hospital, Greece (K.T.); University of Alabama at Birmingham (M.A.L.); Detroit Medical Center, Cardiovascular Institute, MI (C.L.G.); and Citizens Memorial Hospital, Bolivar, MO and University of Missouri, Columbia (K.M.)
| |
Collapse
|
130
|
Yang Y, Bu P. Progress on the cardiotoxicity of sunitinib: Prognostic significance, mechanism and protective therapies. Chem Biol Interact 2016; 257:125-31. [PMID: 27531228 DOI: 10.1016/j.cbi.2016.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/23/2016] [Accepted: 08/05/2016] [Indexed: 12/15/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are multi-targeted anti-cancer agents effective in the treatment of renal cell carcinoma (RCC), imatinib-resistant gastrointestinal stromal tumor (GIST) and pancreatic cancer (PC). Targeting and inhibiting a wide range of oncogenically relevant receptor tyrosine kinases (RTKs), TKIs have been the golden standard treatment of several types of cancer. The cardiotoxicity of TKIs, however, has also emerged alongside their anti-cancer potencies and has attracted research attention. Over the past few years significant progress has been made in developing a deeper understanding of aspects such as extent of cardiotoxicity, prognostic implications and survival predictions, toxicological mechanisms, and potential cardioprotective therapies. In this review we focus on a typical TKI sunitinib and summarize the up-to-date knowledge of sunitinib-induced cardiac abnormalities reported in clinical studies, weighing their implications of prognostic values. We also examine recent findings in underlying mechanisms, and development of potential cardioprotective agents.
Collapse
Affiliation(s)
- Yi Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peili Bu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
131
|
Russell RR, Alexander J, Jain D, Poornima IG, Srivastava AV, Storozynsky E, Schwartz RG. The role and clinical effectiveness of multimodality imaging in the management of cardiac complications of cancer and cancer therapy. J Nucl Cardiol 2016; 23:856-84. [PMID: 27251147 DOI: 10.1007/s12350-016-0538-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 12/24/2022]
Abstract
With the increasing number of individuals living with a current or prior diagnosis of cancer, it is important for the cardiovascular specialist to recognize the various complications of cancer and its therapy on the cardiovascular system. This is true not only for established cancer therapies, such as anthracyclines, that have well established cardiovascular toxicities, but also for the new targeted therapies that can have "off target" effects in the heart and vessels. The purpose of this informational statement is to provide cardiologists, cardiac imaging specialists, cardio-oncologists, and oncologists an understanding of how multimodality imaging may be used in the diagnosis and management of the cardiovascular complications of cancer therapy. In addition, this document is meant to provide useful general information concerning the cardiovascular complications of cancer and cancer therapy as well as established recommendations for the monitoring of specific cardiotoxic therapies.
Collapse
Affiliation(s)
- Raymond R Russell
- Rhode Island Cardiovascular Institute, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 737, Providence, RI, 02903, USA.
| | - Jonathan Alexander
- Cardiology Division, Western Connecticut Medical Center at Danbury Hospital, Danbury, CT, USA
| | - Diwakar Jain
- Section of Cardiovascular Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Indu G Poornima
- Division of Cardiology, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ajay V Srivastava
- Division of Cardiovascular Medicine, Scripps Clinic, La Jolla, CA, USA
| | - Eugene Storozynsky
- Cardiology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Ronald G Schwartz
- Cardiology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Nuclear Medicine Division, Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
132
|
Hinchcliff ME, Lomasney J, Johnson JA, Varga J. Fulminant capillary leak syndrome in a patient with systemic sclerosis treated with imatinib mesylate. Rheumatology (Oxford) 2016; 55:1916-8. [PMID: 27338086 DOI: 10.1093/rheumatology/kew245] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Jon Lomasney
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | |
Collapse
|
133
|
Sridharan V, Thomas CJ, Cao M, Melnyk SB, Pavliv O, Joseph J, Singh SP, Sharma S, Moros EG, Boerma M. Effects of local irradiation combined with sunitinib on early remodeling, mitochondria, and oxidative stress in the rat heart. Radiother Oncol 2016; 119:259-64. [PMID: 27072940 PMCID: PMC4909572 DOI: 10.1016/j.radonc.2016.03.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/23/2016] [Accepted: 03/28/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE Thoracic (chemo)radiation therapy is increasingly administered with tyrosine kinase inhibitors (TKI). While TKI have adverse effects on the heart, it is unknown whether combination with other cancer therapies causes enhanced toxicity. We used an animal model to investigate whether radiation and sunitinib interact in their effects on the heart. MATERIAL AND METHODS Male Sprague-Dawley rats received local heart irradiation (9Gy per day, 5days). Oral sunitinib (8 or 15mg/kg bodyweight per day) started on day 1 of irradiation and continued for 2weeks. Cardiac function was examined with echocardiography. Cardiac remodeling, cell death, left ventricular (LV) oxidative stress markers, mitochondrial morphology and mitochondrial permeability transition pore (mPTP) opening were assessed. RESULTS Cardiac diameter, stroke volume, and LV volume, mass and anterior wall thickness increased in time, but only in the vehicle group. Sunitinib reduced LV inner diameter and volume in systole, which were counteracted by radiation. Sunitinib and radiation showed enhanced effects on mitochondrial morphology and mPTP opening, but not on cardiac troponin I, mast cell numbers or markers of oxidative stress. CONCLUSIONS This study found no early enhanced effects of radiation and sunitinib on cardiac function or structure. Long-term effects remain to be determined.
Collapse
Affiliation(s)
- Vijayalakshmi Sridharan
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, United States
| | | | - Maohua Cao
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, United States
| | - Stepan B Melnyk
- University of Arkansas for Medical Sciences, Department of Pediatrics, Little Rock, United States
| | - Oleksandra Pavliv
- University of Arkansas for Medical Sciences, Department of Pediatrics, Little Rock, United States
| | - Jacob Joseph
- Veterans Affairs Boston Healthcare System, Department of Medicine, United States; Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, United States
| | - Sharda P Singh
- University of Arkansas for Medical Sciences, Department of Pharmacology and Toxicology, Little Rock, United States
| | - Sunil Sharma
- University of Arkansas for Medical Sciences, Department of Radiation Oncology, Little Rock, United States
| | - Eduardo G Moros
- Moffitt Cancer Center and Research Institute, Department of Radiation Oncology, Tampa, United States
| | - Marjan Boerma
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, United States.
| |
Collapse
|
134
|
Mercurio V, Pirozzi F, Lazzarini E, Marone G, Rizzo P, Agnetti G, Tocchetti CG, Ghigo A, Ameri P. Models of Heart Failure Based on the Cardiotoxicity of Anticancer Drugs. J Card Fail 2016; 22:449-58. [PMID: 27103426 DOI: 10.1016/j.cardfail.2016.04.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/13/2022]
Abstract
Heart failure (HF) is a complication of oncological treatments that may have dramatic clinical impact. It may acutely worsen a patient's condition or it may present with delayed onset, even years after treatment, when cancer has been cured or is in stable remission. Several studies have addressed the mechanisms of cancer therapy-related HF and some have led to the definition of disease models that hold valid for other and more common types of HF. Here, we review these models of HF based on the cardiotoxicity of antineoplastic drugs and classify them in cardiomyocyte-intrinsic, paracrine, or potentially secondary to effects on cardiac progenitor cells. The first group includes HF resulting from the combination of oxidative stress, mitochondrial dysfunction, and activation of the DNA damage response, which is typically caused by anthracyclines, and HF resulting from deranged myocardial energetics, such as that triggered by anthracyclines and sunitinib. Blockade of the neuregulin-1/ErbB4/ErbB2, vascular endothelial growth factor/vascular endothelial growth factor receptor and platelet-derived growth factor /platelet-derived growth factor receptor pathways by trastuzumab, sorafenib and sunitinib is proposed as paradigm of cancer therapy-related HF associated with alterations of myocardial paracrine pathways. Finally, anthracyclines and trastuzumab are also presented as examples of antitumor agents that induce HF by affecting the cardiac progenitor cell population.
Collapse
Affiliation(s)
- Valentina Mercurio
- Division of Internal Medicine, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Flora Pirozzi
- Division of Internal Medicine, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Edoardo Lazzarini
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| | - Giancarlo Marone
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Giulio Agnetti
- Johns Hopkins University, Cardiology, Baltimore, Maryland; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Carlo G Tocchetti
- Division of Internal Medicine, Department of Translational Medical Sciences, Federico II University, Naples, Italy.
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Pietro Ameri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genova, Italy
| |
Collapse
|
135
|
Bellinger AM, Arteaga CL, Force T, Humphreys BD, Demetri GD, Druker BJ, Moslehi JJ. Cardio-Oncology: How New Targeted Cancer Therapies and Precision Medicine Can Inform Cardiovascular Discovery. Circulation 2016; 132:2248-58. [PMID: 26644247 DOI: 10.1161/circulationaha.115.010484] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cardio-oncology (the cardiovascular care of cancer patients) has developed as a new translational and clinical field based on the expanding repertoire of mechanism-based cancer therapies. Although these therapies have changed the natural course of many cancers, several may also lead to cardiovascular complications. Many new anticancer drugs approved over the past decade are "targeted" kinase inhibitors that interfere with intracellular signaling contributing to tumor progression. Unexpected cardiovascular and cardiometabolic effects of patient treatment with these inhibitors have provided unique insights into the role of kinases in human cardiovascular biology. Today, an ever-expanding number of cancer therapies targeting novel kinases and other specific cellular and metabolic pathways are being developed and tested in oncology clinical trials. Some of these drugs may affect the cardiovascular system in detrimental ways and others perhaps in beneficial ways. We propose that the numerous ongoing oncology clinical trials are an opportunity for closer collaboration between cardiologists and oncologists to study the cardiovascular and cardiometabolic changes caused by the modulation of these pathways in patients. In this regard, cardio-oncology represents an opportunity and a novel platform for basic and translational investigation and can serve as a potential avenue for optimization of anticancer therapies and for cardiovascular research and drug discovery.
Collapse
Affiliation(s)
- Andrew M Bellinger
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Carlos L Arteaga
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Thomas Force
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Benjamin D Humphreys
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - George D Demetri
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Brian J Druker
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Javid J Moslehi
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.).
| |
Collapse
|
136
|
Morbidelli L, Donnini S, Ziche M. Targeting endothelial cell metabolism for cardio-protection from the toxicity of antitumor agents. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2016; 2:3. [PMID: 33530139 PMCID: PMC7837145 DOI: 10.1186/s40959-016-0010-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/16/2016] [Indexed: 12/17/2022]
Abstract
The vascular endothelium plays a fundamental role in the maintenance of tissue homeostasis, regulating local blood flow and other physiological processes. Chemotherapeutic drugs and target therapies, including antiangiogenic drugs targeting vascular endothelial growth factor (VEGF) or its receptors, not only efficiently act against tumor growth, but may also induce endothelial dysfunction and cardiovascular toxicity. Continued research efforts aim to better understand, prevent and mitigate these chemotherapy associated cardiovascular diseases. Conventional chemotherapeutic agents, such as anthracyclines, platinum compounds, and taxanes, and newer targeted agents, such as bevacizumab, trastuzumab, and tyrosine kinase inhibitors, have known risk of cardiovascular toxicity, which can limit their effectiveness by promoting increased morbidity and/or mortality. This review describes a) the activity of anticancer agents in inducing endothelial dysfunction, b) the metabolic pathways and signalling cascades which may be targeted by protective agents able to maintain or restore endothelial cell function, such as endothelial nitric oxide synthase/fibroblast growth factor-2 (eNOS-FGF-2) pathway, and c) the drugs/strategies reported to improve endothelial function and to reduce the risks of cardiovascular diseases such as angiotensin converting enzyme inhibitors (ACEi) and beta blockers, that are fundamental therapies in chronic heart failure (HF), as well as non-standard HF treatments such ad nitric oxide donors and antioxidant strategies. There is increasing interest in whether ACEi, beta-blockers, and/or statins might prevent and/or therapeutically control cardiotoxic effects in cancer patients. Maintaining endothelial function during or following treatments with chemotherapeutic agents, without affecting anti-tumor drug-effectiveness, is essential for preserving or recovering cardiovascular homeostasis. In this respect, the early detection and immediate therapy of cardiovascular toxicity appear crucial for substantial recovery of cardiac function in cancer patients.
Collapse
Affiliation(s)
- Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Marina Ziche
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
137
|
Zeng H, He X, Tuo QH, Liao DF, Zhang GQ, Chen JX. LPS causes pericyte loss and microvascular dysfunction via disruption of Sirt3/angiopoietins/Tie-2 and HIF-2α/Notch3 pathways. Sci Rep 2016; 6:20931. [PMID: 26868537 PMCID: PMC4751495 DOI: 10.1038/srep20931] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/13/2016] [Indexed: 12/17/2022] Open
Abstract
Recent studies reveal a crucial role of pericyte loss in sepsis-associated microvascular dysfunction. Sirtuin 3 (SIRT3) mediates histone protein post-translational modification related to aging and ischemic disease. This study investigated the involvement of SIRT3 in LPS-induced pericyte loss and microvascular dysfunction. Mice were exposed to LPS, expression of Sirt3, HIF-2α, Notch3 and angiopoietins/Tie-2, pericyte/endothelial (EC) coverage and vascular permeability were assessed. Mice treated with LPS significantly reduced the expression of SIRT3, HIF-2α and Notch3 in the lung. Furthermore, exposure to LPS increased Ang-2 while inhibited Ang-1/Tie-2 expression with a reduced pericyte/EC coverage. Intriguingly, knockout of Sirt3 upregulated Ang-2, but downregulated Tie-2 and HIF-2α/Notch3 expression which resulted in a dramatic reduction of pericyte/EC coverage and exacerbation of LPS-induced vascular leakage. Conversely, overexpression of Sirt3 reduced Ang-2 expression and increased Ang-1/Tie-2 and HIF-2α/Notch3 expression in the LPS treated mice. Overexpression of Sirt3 further prevented LPS-induced pericyte loss and vascular leakage. This was accompanied by a significant reduction of the mortality rate. Specific knockout of prolyl hydroxylase-2 (PHD2) increased HIF-2α/Notch3 expression, improved pericyte/EC coverage and reduced the mortality rate in the LPS-treated mice. Our study demonstrates the importance of SIRT3 in preserving vascular integrity by targeting pericytes in the setting of LPS-induced sepsis.
Collapse
Affiliation(s)
- Heng Zeng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Xiaochen He
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Qin-Hui Tuo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Duan-Fang Liao
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Guo-Qiang Zhang
- Emergency Department of China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| |
Collapse
|
138
|
Eroğlu C, Seçme M, Atmaca P, Kaygusuz O, Gezer K, Bağcı G, Dodurga Y. Extract of Calvatia gigantea inhibits proliferation of A549 human lung cancer cells. Cytotechnology 2016; 68:2075-81. [PMID: 26820971 DOI: 10.1007/s10616-016-9947-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/16/2016] [Indexed: 12/18/2022] Open
Abstract
In this study, in order to investigate the anticancer mechanism of Calvatia gigantea extract, edible mushroom species, which belong to Lycoperdaceae family, changes of CCND1, CCND2, CDK4, p21, Akt, Bax, Bcl-2, p53, caspase-3 and caspase-9 were evaluated in A549 lung cancer cells. Cytotoxic effect of C. gigantea extract was evaluated by using XTT (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5 carboxanilide). The C. gigantea extract was treated in a time and dose dependent manner within the range 25 μg/ml-2 mg/ml to determine the IC50 dose. IC50 dose for C. gigantea extract was detected as 500 μg/ml for 72 h. According to expression results, while CCND1, CCND2, CDK4, Akt and Bcl-2 expression clearly decreased, Bax, p53, caspase-3 and caspase-9 expression clearly increased in the dose group cells (A549 cells treated with 500 μg/ml dose of C. gigantea extract for 72 h). However, there was no change in p21 expression. C. gigantea extract induced cell cycle arrest and apoptosis by decreasing the CCND1, CCND2, CDK4, Akt and Bcl-2 expression and by increasing Bax, p53, caspase-3 and caspase-9 expression in A549 cells. Mushrooms are eukaryotic organisms heavily used because of their supposedly anticancer effect. Many mushroom species have been used for medical purposes, as a result of also having many effects such as antibiotic, antiviral and anticancer effects. It is thought that the C. gigantea extract may be a significant agent for treatment of lung cancer as a single agent or in combination with other drugs.
Collapse
Affiliation(s)
- Canan Eroğlu
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Kınıklı/Denizli, Turkey.
| | - Mücahit Seçme
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Kınıklı/Denizli, Turkey
| | - Pelin Atmaca
- Department of Biology, Faculty of Science, Pamukkale University, Denizli, Turkey
| | - Oğuzhan Kaygusuz
- Department of Biology, Faculty of Science, Pamukkale University, Denizli, Turkey
| | - Kutret Gezer
- Department of Biology, Faculty of Science, Pamukkale University, Denizli, Turkey
| | - Gülseren Bağcı
- Department of Medical Genetics, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Yavuz Dodurga
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Kınıklı/Denizli, Turkey
| |
Collapse
|
139
|
Iliescu CA, Grines CL, Herrmann J, Yang EH, Cilingiroglu M, Charitakis K, Hakeem A, Toutouzas KP, Leesar MA, Marmagkiolis K. SCAI Expert consensus statement: Evaluation, management, and special considerations of cardio-oncology patients in the cardiac catheterization laboratory (endorsed by the cardiological society of india, and sociedad Latino Americana de Cardiologıa intervencionista). Catheter Cardiovasc Interv 2016; 87:E202-23. [PMID: 26756277 DOI: 10.1002/ccd.26379] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/28/2015] [Indexed: 12/24/2022]
Abstract
In the United States alone, there are currently approximately 14.5 million cancer survivors, and this number is expected to increase to 20 million by 2020. Cancer therapies can cause significant injury to the vasculature, resulting in angina, acute coronary syndromes (ACS), stroke, critical limb ischemia, arrhythmias, and heart failure, independently from the direct myocardial or pericardial damage from the malignancy itself. Consequently, the need for invasive evaluation and management in the cardiac catheterization laboratory (CCL) for such patients has been increasing. In recognition of the need for a document on special considerations for cancer patients in the CCL, the Society for Cardiovascular Angiography and Interventions (SCAI) commissioned a consensus group to provide recommendations based on the published medical literature and on the expertise of operators with accumulated experience in the cardiac catheterization of cancer patients.
Collapse
Affiliation(s)
- Cezar A Iliescu
- MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Cindy L Grines
- Detroit Medical Center, Cardiovascular Institute, Detroit, Michigan
| | - Joerg Herrmann
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Eric H Yang
- Division of Cardiology, University of California at Los Angeles, Los Angeles, California
| | - Mehmet Cilingiroglu
- School of Medicine, Arkansas Heart Hospital, Little Rock, Arkansas.,Department of Cardiology, Koc University, Istanbul, Turkey
| | | | - Abdul Hakeem
- Department of Cardiology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Massoud A Leesar
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Konstantinos Marmagkiolis
- Department of Cardiology, Citizens Memorial Hospital, Bolivar, Missouri.,Department of Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
140
|
Argyros O, Karampelas T, Asvos X, Varela A, Sayyad N, Papakyriakou A, Davos CH, Tzakos AG, Fokas D, Tamvakopoulos C. Peptide–Drug Conjugate GnRH–Sunitinib Targets Angiogenesis Selectively at the Site of Action to Inhibit Tumor Growth. Cancer Res 2015; 76:1181-92. [DOI: 10.1158/0008-5472.can-15-2138] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 11/24/2015] [Indexed: 11/16/2022]
|
141
|
Kreuger J, Phillipson M. Targeting vascular and leukocyte communication in angiogenesis, inflammation and fibrosis. Nat Rev Drug Discov 2015; 15:125-42. [PMID: 26612664 DOI: 10.1038/nrd.2015.2] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regulation of vascular permeability, recruitment of leukocytes from blood to tissue and angiogenesis are all processes that occur at the level of the microvasculature during both physiological and pathological conditions. The interplay between microvascular cells and leukocytes during inflammation, together with the emerging roles of leukocytes in the modulation of the angiogenic process, make leukocyte-vascular interactions prime targets for therapeutics to potentially treat a wide range of diseases, including pathological and dysfunctional vessel growth, chronic inflammation and fibrosis. In this Review, we discuss how the different cell types that are present in and around microvessels interact, cooperate and instruct each other, and in this context we highlight drug targets as well as emerging druggable processes that can be exploited to restore tissue homeostasis.
Collapse
Affiliation(s)
- Johan Kreuger
- Department of Medical Cell Biology, Uppsala University, Husargatan 3, Uppsala, 75123, Sweden
| | - Mia Phillipson
- Department of Medical Cell Biology, Uppsala University, Husargatan 3, Uppsala, 75123, Sweden
| |
Collapse
|
142
|
Chen WCW, Baily JE, Corselli M, Díaz ME, Sun B, Xiang G, Gray GA, Huard J, Péault B. Human myocardial pericytes: multipotent mesodermal precursors exhibiting cardiac specificity. Stem Cells 2015; 33:557-73. [PMID: 25336400 DOI: 10.1002/stem.1868] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 09/08/2014] [Accepted: 09/29/2014] [Indexed: 12/20/2022]
Abstract
Perivascular mesenchymal precursor cells (i.e., pericytes) reside in skeletal muscle where they contribute to myofiber regeneration; however, the existence of similar microvessel-associated regenerative precursor cells in cardiac muscle has not yet been documented. We tested whether microvascular pericytes within human myocardium exhibit phenotypes and multipotency similar to their anatomically and developmentally distinct counterparts. Fetal and adult human heart pericytes (hHPs) express canonical pericyte markers in situ, including CD146, NG2, platelet-derived growth factor receptor (PDGFR) β, PDGFRα, alpha-smooth muscle actin, and smooth muscle myosin heavy chain, but not CD117, CD133, and desmin, nor endothelial cell (EC) markers. hHPs were prospectively purified to homogeneity from ventricular myocardium by flow cytometry, based on a combination of positive- (CD146) and negative-selection (CD34, CD45, CD56, and CD117) cell lineage markers. Purified hHPs expanded in vitro were phenotypically similar to human skeletal muscle-derived pericytes (hSkMPs). hHPs express mesenchymal stem/stromal cell markers in situ and exhibited osteo-, chondro-, and adipogenic potentials but, importantly, no ability for skeletal myogenesis, diverging from pericytes of all other origins. hHPs supported network formation with/without ECs in Matrigel cultures; hHPs further stimulated angiogenic responses under hypoxia, markedly different from hSkMPs. The cardiomyogenic potential of hHPs was examined following 5-azacytidine treatment and neonatal cardiomyocyte coculture in vitro, and intramyocardial transplantation in vivo. Results indicated cardiomyocytic differentiation in a small fraction of hHPs. In conclusion, human myocardial pericytes share certain phenotypic and developmental similarities with their skeletal muscle homologs, yet exhibit different antigenic, myogenic, and angiogenic properties. This is the first example of an anatomical restriction in the developmental potential of pericytes as native mesenchymal stem cells.
Collapse
Affiliation(s)
- William C W Chen
- Department of Bioengineering, University of Pittsburgh, Pennsylvania, USA; Department of Orthopedic Surgery, University of Pittsburgh, Pennsylvania, USA; Stem Cell Research Centre, University of Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Traumatic brain injury results in rapid pericyte loss followed by reactive pericytosis in the cerebral cortex. Sci Rep 2015; 5:13497. [PMID: 26333872 PMCID: PMC4558600 DOI: 10.1038/srep13497] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/28/2015] [Indexed: 01/08/2023] Open
Abstract
Accumulating evidence suggests a pivotal role of PDGFRß positive cells, a specific marker for central nervous system (CNS) pericytes, in tissue scarring. Identification of cells that contribute to tissue reorganization in the CNS upon injury is a crucial step to develop novel treatment strategies in regenerative medicine. It has been shown that pericytes contribute to scar formation in the spinal cord. It is further known that ischemia initially triggers pericyte loss in vivo, whilst brain trauma is capable of inducing pericyte detachment from cerebral vessels. These data point towards a significant role of pericytes in CNS injury. The temporal and spatial dynamics of PDGFRß cells and their responses in traumatic brain injury are poorly understood. Here we show that PDGFRß positive cells initially decline in the acute phase following experimental traumatic brain injury. However, PDGFRß positive cells increase significantly in the trauma zone days after brain injury. Using various pericyte markers we identify these cells to be pericytes that are demarcated by reactive gliosis. Our data indicate that brain trauma causes a biphasic response of pericytes in the early phase of brain trauma that may be of relevance for the understanding of pathological cellular responses in traumatic brain injury.
Collapse
|
144
|
Kennedy-Lydon T, Crawford C, Wildman SS, Peppiatt-Wildman CM. Nonsteroidal anti-inflammatory drugs alter vasa recta diameter via pericytes. Am J Physiol Renal Physiol 2015. [PMID: 26202223 DOI: 10.1152/ajprenal.00199.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We have previously shown that vasa recta pericytes are known to dilate vasa recta capillaries in the presence of PGE2 and contract vasa recta capillaries when endogenous production of PGE2 is inhibited by the nonselective nonsteroidal anti-inflammatory drug (NSAID) indomethacin. In the present study, we used a live rat kidney slice model to build on these initial observations and provide novel data that demonstrate that nonselective, cyclooxygenase-1-selective, and cyclooxygenase -2-selective NSAIDs act via medullary pericytes to elicit a reduction of vasa recta diameter. Real-time images of in situ vasa recta were recorded, and vasa recta diameters at pericyte and nonpericyte sites were measured offline. PGE2 and epoprostenol (a prostacyclin analog) evoked dilation of vasa recta specifically at pericyte sites, and PGE2 significantly attenuated pericyte-mediated constriction of vasa recta evoked by both endothelin-1 and ANG II. NSAIDs (indomethacin > SC-560 > celecoxib > meloxicam) evoked significantly greater constriction of vasa recta capillaries at pericyte sites than at nonpericyte sites, and indomethacin significantly attenuated the pericyte-mediated vasodilation of vasa recta evoked by PGE2, epoprostenol, bradykinin, and S-nitroso-N-acetyl-l-penicillamine. Moreover, a reduction in PGE2 was measured using an enzyme immune assay after superfusion of kidney slices with indomethacin. In addition, immunohistochemical techniques were used to demonstrate the population of EP receptors in the medulla. Collectively, these data demonstrate that pericytes are sensitive to changes in PGE2 concentration and may serve as the primary mechanism underlying NSAID-associated renal injury and/or further compound-associated tubular damage.
Collapse
Affiliation(s)
- Teresa Kennedy-Lydon
- Urinary System Physiology Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Kent, United Kingdom
| | - Carol Crawford
- Urinary System Physiology Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Kent, United Kingdom
| | - Scott S Wildman
- Urinary System Physiology Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Kent, United Kingdom
| | - Claire M Peppiatt-Wildman
- Urinary System Physiology Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Kent, United Kingdom
| |
Collapse
|
145
|
Retrospective analysis of the safety and efficacy of high-dose interleukin-2 after prior tyrosine kinase inhibitor therapy in patients with advanced renal cell carcinoma. J Immunother 2015; 37:360-5. [PMID: 25075565 PMCID: PMC4127096 DOI: 10.1097/cji.0000000000000044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Although tyrosine kinase inhibitors (TKI) are the most common first-line therapy for metastatic renal cell carcinoma, high-dose interleukin-2 (HD-IL2) remains the only agent that provides durable complete responses. The optimal sequence of these agents remains uncertain. This retrospective multi-institutional study examined the safety and efficacy of HD-IL2 following TKI therapy. After IRB approval at 7 HD-IL2 centers, data relating to patient, disease, and treatment characteristics among 40 consecutive patients with metastatic renal cell carcinoma who were treated with HD-IL2 after at least 1 prior TKI therapy were retrospectively collected. The most common cardiac adverse events were grade 3 hypotension and vascular leak syndrome. Six patients (15%) experienced other grade ≥3 cardiac adverse events. There were 2 treatment-related deaths due to congestive heart failure, occurring in 1 patient with short TKI to HD-IL2 interval and another patient with an abnormal baseline cardiac stress test. Best responses included 2 CRs (5%, duration 40+ and 62+ mo), 3 PRs (8%, duration 6, 11, and 24 mo), 13 SD (32%, median duration 12 mo), 20 PD (50%), and 2 not evaluable patients. Median overall survival was 22 months. Administration of HD-IL2 could be safe and effective after TKI therapy; however, careful selection of patients is critical. We recommend baseline cardiac risk factor assessment, screening with both cardiac stress test and echocardiogram, and allowing a TKI to HD-IL2 interval of at least 2 months.
Collapse
|
146
|
Rees ML, Subramaniam J, Li Y, Hamilton DJ, Frazier OH, Taegtmeyer H. A PKM2 signature in the failing heart. Biochem Biophys Res Commun 2015; 459:430-6. [PMID: 25735978 PMCID: PMC4380635 DOI: 10.1016/j.bbrc.2015.02.122] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/21/2015] [Indexed: 11/28/2022]
Abstract
A salient feature of the failing heart is metabolic remodeling towards predominant glucose metabolism and activation of the fetal gene program. Sunitinib is a multitargeted receptor tyrosine kinase inhibitor used for the treatment of highly vascularized tumors. In diabetic patients, sunitinib significantly decreases blood glucose. However, a considerable proportion of sunitinib-treated patients develop cardiac dysfunction or failure. We asked whether sunitinib treatment results in shift towards glycolysis in the heart. Glucose uptake by the heart was increased fivefold in mice treated with sunitinib. Transcript analysis by qPCR revealed an induction of genes associated with glycolysis and reactivation of the fetal gene program. Additionally, we observed a shift in the enzyme pyruvate kinase from the adult M1 (PKM1) isoform to the fetal M2 (PKM2) isoform, a hallmark of the Warburg Effect. This novel observation led us to examine whether a similar shift occurs in human heart failure. Examination of tissue from patients with heart failure similarly displayed an induction of PKM2. Moreover, this phenomenon was partially reversed following mechanical unloading. We propose that pyruvate kinase isoform switching represents a novel feature of the fetal gene program in the failing heart.
Collapse
Affiliation(s)
- Meredith L Rees
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA
| | - Janani Subramaniam
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA
| | - Yuanteng Li
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA
| | - Dale J Hamilton
- Department of Medicine, Division of Endocrinology, Bioenergetic Laboratory, Houston Methodist Research Institute, 6550 Fannin Street, #1001, Houston, TX 77030, USA
| | - O Howard Frazier
- Texas Heart Institute, CHI St. Luke's Health - Baylor St. Luke's Medical Center, MC 2-114A, PO Box 20345, Houston, TX 77225, USA
| | - Heinrich Taegtmeyer
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA; Texas Heart Institute, CHI St. Luke's Health - Baylor St. Luke's Medical Center, MC 2-114A, PO Box 20345, Houston, TX 77225, USA.
| |
Collapse
|
147
|
Trembley MA, Velasquez LS, de Mesy Bentley KL, Small EM. Myocardin-related transcription factors control the motility of epicardium-derived cells and the maturation of coronary vessels. Development 2015; 142:21-30. [PMID: 25516967 DOI: 10.1242/dev.116418] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
An important pool of cardiovascular progenitor cells arises from the epicardium, a single layer of mesothelium lining the heart. Epicardium-derived progenitor cell (EPDC) formation requires epithelial-to-mesenchymal transition (EMT) and the subsequent migration of these cells into the sub-epicardial space. Although some of the physiological signals that promote EMT are understood, the functional mediators of EPDC motility and differentiation are not known. Here, we identify a novel regulatory mechanism of EPDC mobilization. Myocardin-related transcription factor (MRTF)-A and MRTF-B (MKL1 and MKL2, respectively) are enriched in the perinuclear space of epicardial cells during development. Transforming growth factor (TGF)-β signaling and disassembly of cell contacts leads to nuclear accumulation of MRTFs and the activation of the motile gene expression program. Conditional ablation of Mrtfa and Mrtfb specifically in the epicardium disrupts cell migration and leads to sub-epicardial hemorrhage, partially stemming from the depletion of coronary pericytes. Using lineage-tracing analyses, we demonstrate that sub-epicardial pericytes arise from EPDCs in a process that requires the MRTF-dependent motile gene expression program. These findings provide novel mechanisms linking EPDC motility and differentiation, shed light on the transcriptional control of coronary microvascular maturation and suggest novel therapeutic strategies to manipulate epicardium-derived progenitor cells for cardiac repair.
Collapse
Affiliation(s)
- Michael A Trembley
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Lissette S Velasquez
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Karen L de Mesy Bentley
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Eric M Small
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| |
Collapse
|
148
|
Cross MJ, Berridge BR, Clements PJM, Cove-Smith L, Force TL, Hoffmann P, Holbrook M, Lyon AR, Mellor HR, Norris AA, Pirmohamed M, Tugwood JD, Sidaway JE, Park BK. Physiological, pharmacological and toxicological considerations of drug-induced structural cardiac injury. Br J Pharmacol 2015; 172:957-74. [PMID: 25302413 PMCID: PMC4314188 DOI: 10.1111/bph.12979] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/30/2014] [Accepted: 10/06/2014] [Indexed: 01/01/2023] Open
Abstract
The incidence of drug-induced structural cardiotoxicity, which may lead to heart failure, has been recognized in association with the use of anthracycline anti-cancer drugs for many years, but has also been shown to occur following treatment with the new generation of targeted anti-cancer agents that inhibit one or more receptor or non-receptor tyrosine kinases, serine/threonine kinases as well as several classes of non-oncology agents. A workshop organized by the Medical Research Council Centre for Drug Safety Science (University of Liverpool) on 5 September 2013 and attended by industry, academia and regulatory representatives, was designed to gain a better understanding of the gaps in the field of structural cardiotoxicity that can be addressed through collaborative efforts. Specific recommendations from the workshop for future collaborative activities included: greater efforts to identify predictive (i) preclinical; and (ii) clinical biomarkers of early cardiovascular injury; (iii) improved understanding of comparative physiology/pathophysiology and the clinical predictivity of current preclinical in vivo models; (iv) the identification and use of a set of cardiotoxic reference compounds for comparative profiling in improved animal and human cellular models; (v) more sharing of data (through publication/consortia arrangements) on target-related toxicities; (vi) strategies to develop cardio-protective agents; and (vii) closer interactions between preclinical scientists and clinicians to help ensure best translational efforts.
Collapse
Affiliation(s)
- M J Cross
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| | - B R Berridge
- Safety Assessment, GlaxoSmithKlineResearch Triangle Park, NC, USA
| | - P J M Clements
- David Jack Centre for Research & Development, GlaxoSmithKlineWare, Herts, UK
| | - L Cove-Smith
- Clinical & Experimental Pharmacology, Cancer Research UK Manchester Institute, University of ManchesterManchester, UK
| | - T L Force
- Center for Translational Medicine and Cardiology Division, Temple University School of MedicinePhiladelphia, PA, USA
| | - P Hoffmann
- Preclinical Safety, Novartis Pharm CorpEast Hanover, NJ, USA
| | - M Holbrook
- Safety Pharmacology, Covance Laboratories, Ltd.Harrogate, North Yorkshire, UK
| | - A R Lyon
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and Imperial CollegeLondon, UK
| | - H R Mellor
- Drug Safety Evaluation, Vertex Pharmaceuticals (Europe), Ltd.Abingdon, Oxfordshire, UK
| | - A A Norris
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| | - M Pirmohamed
- The Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of LiverpoolLiverpool, UK
| | - J D Tugwood
- Clinical & Experimental Pharmacology, Cancer Research UK Manchester Institute, University of ManchesterManchester, UK
| | - J E Sidaway
- Innovative Medicines, AstraZeneca R&DMacclesfield, UK
| | - B K Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| |
Collapse
|
149
|
Bronte G, Bronte E, Novo G, Pernice G, Lo Vullo F, Musso E, Bronte F, Gulotta E, Rizzo S, Rolfo C, Silvestris N, Bazan V, Novo S, Russo A. Conquests and perspectives of cardio-oncology in the field of tumor angiogenesis-targeting tyrosine kinase inhibitor-based therapy. Expert Opin Drug Saf 2014; 14:253-67. [PMID: 25494575 DOI: 10.1517/14740338.2015.986092] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Angiogenesis is fundamental for tumor development and progression. Hence, anti-angiogenic drugs have been developed to target VEGF and its receptors (VEGFRs). Several tyrosine kinase inhibitors (TKIs) have been developed over the years and others are still under investigation, each anti-VEGFR TKI showing a different cardiotoxic profile. Knowledge of the cardiac side-effects of each drug and the magnitude of their expression and frequency can lead to a specific approach. AREAS COVERED This work reviews the mechanism of action of anti-VEGFR TKIs and the pathophysiological mechanisms leading to cardiotoxicity, followed by close examination of the most important drugs individually. A literature search was conducted on PubMed selecting review articles, original studies and clinical trials, with a focus on Phase III studies. EXPERT OPINION Side-effects on the cardiovascular system could lead both to the worsening of general health status of cancer patients and to the discontinuation of the cancer treatment affecting its efficacy. Cardiologists often have to face new triggers of heart disease in these patients. They need a specific approach, which must be carried out in cooperation with oncologists. It must start before cancer treatment, continue during it and extend after its completion.
Collapse
Affiliation(s)
- Giuseppe Bronte
- University of Palermo, Department of Surgical, Oncological and Oral Sciences , Palermo , Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Recognizing and managing left ventricular dysfunction associated with therapeutic inhibition of the vascular endothelial growth factor signaling pathway. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2014; 16:335. [PMID: 25099086 DOI: 10.1007/s11936-014-0335-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OPINION STATEMENT Therapeutic inhibition of the vascular endothelial growth factor (VEGF) signaling pathway (VSP) is increasingly employed in the contemporary treatment of many cancer types. VSP inhibitors include the anti-VEGF monoclonal antibody (bevacizumab), soluble VEGF receptors (VEGF Trap), and small molecule tyrosine kinase inhibitors (TKIs) targeting the intracellular kinase domain of VEGF receptors. These agents are associated with cardiovascular toxicities such as hypertension, thrombosis, myocardial ischemia, and left ventricular (LV) dysfunction. Data on VSP inhibitor-associated LV dysfunction are largely limited to retrospective studies. Prospective studies are needed to establish the clinical significance of VSP inhibitor-associated LV dysfunction in the general population. Pre-clinical models of VSP inhibitor-associated LV dysfunction have identified mechanisms of cardiotoxicity and may improve our understanding of the pathophysiology underlying other cardiomyopathies. This review provides an overview of LV dysfunction that can occur in patients treated with VSP inhibitors. Potential strategies for clinical detection and management of this cardiotoxicity are explored, while acknowledging that currently available data specific to VSP-inhibitor LV dysfunction are limited. Avenues for future research are suggested.
Collapse
|