1
|
Bisceglia I, Venturini E, Canale ML, Ambrosetti M, Riccio C, Giallauria F, Gallucci G, Abrignani MG, Russo G, Lestuzzi C, Mistrulli R, De Luca G, Maria Turazza F, Mureddu G, Di Fusco SA, Lucà F, De Luca L, Camerini A, Halasz G, Camilli M, Quagliariello V, Maurea N, Fattirolli F, Gulizia MM, Gabrielli D, Grimaldi M, Colivicchi F, Oliva F. Cardio-oncology rehabilitation: are we ready? Eur Heart J Suppl 2024; 26:ii252-ii263. [PMID: 38784673 PMCID: PMC11110456 DOI: 10.1093/eurheartjsupp/suae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Cardio-oncology rehabilitation (CORE) is not only an essential component of cancer rehabilitation but also a pillar of preventive cardio-oncology. Cardio-oncology rehabilitation is a comprehensive model based on a multitargeted approach and its efficacy has been widely documented; when compared with an 'exercise only' programme, comprehensive CORE demonstrates a better outcome. It involves nutritional counselling, psychological support, and cardiovascular (CV) risk assessment, and it is directed to a very demanding population with a heavy burden of CV diseases driven by physical inactivity, cancer therapy-induced metabolic derangements, and cancer therapy-related CV toxicities. Despite its usefulness, CORE is still underused in cancer patients and we are still at the dawning of remote models of rehabilitation (tele-rehabilitation). Not all CORE is created equally: a careful screening procedure to identify patients who will benefit the most from CORE and a multidisciplinary customized approach are mandatory to achieve a better outcome for cancer survivors throughout their cancer journey. The aim of this paper is to provide an updated review of CORE not only for cardiologists dealing with this peculiar population of patients but also for oncologists, primary care providers, patients, and caregivers. This multidisciplinary team should help cancer patients to maintain a healthy and active life before, during, and after cancer treatment, in order to improve quality of life and to fight health inequities.
Collapse
Affiliation(s)
- Irma Bisceglia
- Integrated Cardiology Services, Cardio-Thoracic-Vascular Department, Azienda Ospedaliera San Camillo Forlanini, C.ne Gianicolense, 87 00152 Rome, Italy
| | - Elio Venturini
- Department of Cardiac Rehabilitation, Cecina Civil Hospital, Via Montanara, 52, 57023 Cecina (LI), Italy
| | - Maria Laura Canale
- Division of Cardiology, Azienda USL Toscana Nord-Ovest, Versilia Hospital, Via Aurelia, 335, 55041 Lido di Camaiore (LU), Italy
| | - Marco Ambrosetti
- Unità Operativa Complessa di Riabilitazione Cardiologica, ASST Crema Ospedale Santa Marta, Rivolta D'Adda, 26027 Cremona, Italy
| | - Carmine Riccio
- Dipartimento Cardio-Vascolare, AORN Sant'Anna e San Sebastiano, Caserta, Italy
| | - Francesco Giallauria
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | | | | | - Giulia Russo
- SC Patologie Cardiovascolari, Ospedale Maggiore, Via Slataper, 9, 34125 Trieste, Italy
| | | | - Raffaella Mistrulli
- Cardiology Unit, Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Giovanni De Luca
- Health Activities and Epidemiologic Observatory Division, Health Department, Sicily Region, Piazza O. Ziino, 24, 90145 Palermo, Italy
| | - Fabio Maria Turazza
- Director of Cardiology, IRCCS Foundation, National Cancer Institute, via G Venezian, 1, 20133 Milano, Italy
| | - Gianfrancesco Mureddu
- Cardiology Division, S. Giovanni Hospital, Via dell'Amba Aradam, 8, 00184 Rome, Italy
| | - Stefania Angela Di Fusco
- Clinical and Rehabilitation Cardiology Unit, San Filippo Neri Hospital, Via Martinotti, 20, 00135 Roma, Italy
| | - Fabiana Lucà
- Cardiology Department, Grande Ospedale Metropolitano, GOM, AO Bianchi Melacrino Morelli, 89129 Reggio di Calabria, Italy
| | - Leonardo De Luca
- SC Cardiologia, Fondazione IRCCS San Matteo, Viale Camillo Golgi, 19, 27100 Pavia, Italy
| | - Andrea Camerini
- Department of Medical Oncology, Azienda USL Toscana Nord-Ovest, Versilia Hospital,Via Aurelia, 335, 55041 Lido di Camaiore (LU), Italy
| | - Geza Halasz
- Dipartimento Cardio-Toraco-Vascolare, U.O.C. Cardiologia, Azienda Ospedaliera San Camillo-Forlanini, C.ne Gianicolense, 87, 00152 Rome, Italy
| | - Massimiliano Camilli
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli, IRCCS, L.go A. Gemelli, 1, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, L.go Francesco Vito, 00168 Rome, Italy
| | - Vincenzo Quagliariello
- Division of Cardiology, National Cancer Institute, Sen. Pascale Foundation, via Mariano Semmola, 80131 Napoli, Italy
| | - Nicola Maurea
- Division of Cardiology, National Cancer Institute, Sen. Pascale Foundation, via Mariano Semmola, 80131 Napoli, Italy
| | - Francesco Fattirolli
- Azienda Ospedaliero - Universitaria Careggi, Largo Brambilla, 3, 50134 Firenze, Italy
| | | | - Domenico Gabrielli
- Dipartimento Cardio-Toraco-Vascolare, U.O.C. Cardiologia, Azienda Ospedaliera San Camillo-Forlanini, C.ne Gianicolense, 87, 00152 Rome, Italy
- Fondazione per il Tuo cuore—Heart Care Foundation, Firenze, Via A. La Marmora, 36, 50121 Firenze, Italy
| | - Massimo Grimaldi
- Regional General Hospital F. Miulli, Strada Prov. 127 Acquaviva – Santeramo Km, 4, 100.70021 Acquaviva delle Fonti (BARI), Italy
| | - Furio Colivicchi
- Clinical and Rehabilitation Cardiology Unit, San Filippo Neri Hospital, Via Martinotti, 20, 00135 Roma, Italy
| | - Fabrizio Oliva
- Cardiologia 1-Emodinamica Dipartimento Cardiotoracovascolare ‘A. De Gasperis’, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| |
Collapse
|
2
|
Screening for Coronary Artery Disease in Cancer Survivors: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2023; 5:22-38. [PMID: 36875910 PMCID: PMC9982229 DOI: 10.1016/j.jaccao.2022.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 02/24/2023] Open
Abstract
Coronary artery disease (CAD) is an important contributor to the cardiovascular burden in cancer survivors. This review identifies features that could help guide decisions about the benefit of screening to assess the risk or presence of subclinical CAD. Screening may be appropriate in selected survivors based on risk factors and inflammatory burden. In cancer survivors who have undergone genetic testing, polygenic risk scores and clonal hematopoiesis markers may become useful CAD risk prediction tools in the future. The type of cancer (especially breast, hematological, gastrointestinal, and genitourinary) and the nature of treatment (radiotherapy, platinum agents, fluorouracil, hormonal therapy, tyrosine kinase inhibitors, endothelial growth factor inhibitors, and immune checkpoint inhibitors) are also important in determining risk. Therapeutic implications of positive screening include lifestyle and atherosclerosis interventions, and in specific instances, revascularization may be indicated.
Collapse
Key Words
- ACS, acute coronary syndrome
- AYA, adolescent and young adult
- CAC, coronary artery calcium
- CAD, coronary artery disease
- CHIP, clonal hematopoiesis of indeterminate potential
- CMR, cardiac magnetic resonance
- CTA, computed tomography angiography
- CVD, cardiovascular disease
- IGF, insulin-like growth factor
- LDL, low-density lipoprotein
- PCE, pooled cohort equations
- PCI, percutaneous coronary intervention
- PRS, polygenic risk score
- ROS, reactive oxygen species
- TKI, tyrosine kinase inhibitor
- VEGF, vascular endothelial growth factor
- calcification
- coronary artery calcium
- coronary artery disease
- prevention
- risk factor
- risk prediction
Collapse
|
3
|
Toro C, Felmingham B, Jessop S, Celermajer DS, Kotecha RS, Govender D, Terese Hanna DM, O'Connor M, Manudhane R, Ayer J, O'Sullivan J, Sullivan M, Costello B, La Gerche A, Walwyn T, Horvath L, Mateos MK, Fulbright J, Jadhav M, Cheung M, Eisenstat D, Elliott DA, Conyers R. Cardio-Oncology Recommendations for Pediatric Oncology Patients: An Australian and New Zealand Delphi Consensus. JACC. ADVANCES 2022; 1:100155. [PMID: 38939459 PMCID: PMC11198111 DOI: 10.1016/j.jacadv.2022.100155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/07/2022] [Accepted: 10/25/2022] [Indexed: 06/29/2024]
Abstract
Cardio-oncology is a new multidisciplinary area of expertise that seeks to pre-emptively and proactively address cardiac complications that emerge during and following cancer therapy. Modern therapies including molecular targeted therapy and immunotherapy have broadened the agents that can cause cardiac sequelae, often with complications arising within days to weeks of therapy. Several international guidelines have been developed for the acute monitoring of cardio-oncology side effects. However, none are specific to pediatrics. We have addressed this gap in the literature by undertaking a rigorous Delphi consensus approach across 11 domains of cardio-oncology care using an Australian and New Zealand expert group. The expert group consisted of pediatric and adult cardiologists and pediatric oncologists. This Delphi consensus provides an approach to perform risk and baseline assessment, screening, and follow-up, specific to the cancer therapeutic. This review is a useful tool for clinicians involved in the cardio-oncology care of pediatric oncology patients.
Collapse
Affiliation(s)
- Claudia Toro
- Cardiac Regeneration Laboratory, Murdoch Children’s Research Institute, Parkville, Melbourne, Australia
- Children’s Cancer Centre, The Royal Children’s Hospital, Parkville, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Ben Felmingham
- Cardiac Regeneration Laboratory, Murdoch Children’s Research Institute, Parkville, Melbourne, Australia
- Children’s Cancer Centre, The Royal Children’s Hospital, Parkville, Melbourne, Australia
| | - Sophie Jessop
- Michael Rice Centre for Haematology and Oncology, Women’s and Children’s Hospital, Adelaide, South Australia, Australia
| | - David S. Celermajer
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiology, RPA Hospital, Camperdown, New South Wales, Australia
| | - Rishi S. Kotecha
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children’s Hospital, Perth, Australia
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia
- Curtin Medical School, Curtin University, Perth, Australia
| | - Dinisha Govender
- Cancer Centre for Children, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Diane Marie Terese Hanna
- Children’s Cancer Centre, The Royal Children’s Hospital, Parkville, Melbourne, Australia
- Murdoch Children's Research Institute, Melbourne University, Parkville, Victoria, Australia
- The Walter & Eliza Hall Institute, Parkville, Victoria, Australia
| | - Matthew O'Connor
- Michael Rice Centre for Haematology and Oncology, Women’s and Children’s Hospital, Adelaide, South Australia, Australia
| | - Rebecca Manudhane
- Michael Rice Centre for Haematology and Oncology, Women’s and Children’s Hospital, Adelaide, South Australia, Australia
| | - Julian Ayer
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- The Heart Centre for Children, The Sydney Children’s Hospital Network Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | - John O'Sullivan
- Department of Cardiology, RPA Hospital, Camperdown, New South Wales, Australia
- Heart Institute, Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Michael Sullivan
- Children’s Cancer Centre, The Royal Children’s Hospital, Parkville, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Ben Costello
- Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - André La Gerche
- Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Thomas Walwyn
- Department of Paediatric Oncology, Haematology and Bone Marrow Transplantation, Perth Children’s Hospital, Nedlands, Western Australia, Australia
- Discipline of Paediatrics, Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Lisa Horvath
- Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Marion K. Mateos
- Kids Cancer Centre, Sydney Children’s Hospital Randwick, Sydney, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, New South Wales, Australia
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - Joy Fulbright
- Division of Pediatric Hematology/Oncology, Children’s Mercy Kansas City, Kansas City, Missouri, USA
| | - Mangesh Jadhav
- Cardiology Department, The Royal Children’s Hospital, Melbourne, Australia
| | - Michael Cheung
- Cardiac Regeneration Laboratory, Murdoch Children’s Research Institute, Parkville, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Australia
- Cardiology Department, The Royal Children’s Hospital, Melbourne, Australia
| | - David Eisenstat
- Children’s Cancer Centre, The Royal Children’s Hospital, Parkville, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - David A. Elliott
- Cardiac Regeneration Laboratory, Murdoch Children’s Research Institute, Parkville, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Rachel Conyers
- Cardiac Regeneration Laboratory, Murdoch Children’s Research Institute, Parkville, Melbourne, Australia
- Children’s Cancer Centre, The Royal Children’s Hospital, Parkville, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Australia
| |
Collapse
|
4
|
Liu J, Chen ZZ, Patel J, Asnani A. Understanding Myocardial Metabolism in the Context of Cardio-Oncology. Heart Fail Clin 2022; 18:415-424. [PMID: 35718416 DOI: 10.1016/j.hfc.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cardiovascular events, ranging from arrhythmias to decompensated heart failure, are common during and after cancer therapy. Cardiovascular complications can be life-threatening, and from the oncologist's perspective, could limit the use of first-line cancer therapeutics. Moreover, an aging population increases the risk for comorbidities and medical complexity among patients who undergo cancer therapy. Many have established cardiovascular diagnoses or risk factors before starting these therapies. Therefore, it is essential to understand the molecular mechanisms that drive cardiovascular events in patients with cancer and to identify new therapeutic targets that may prevent and treat these 2 diseases. This review will discuss the metabolic interaction between cancer and the heart and will highlight current strategies of targeting metabolic pathways for cancer treatment. Finally, this review highlights opportunities and challenges in advancing our understanding of myocardial metabolism in the context of cancer and cancer treatment.
Collapse
Affiliation(s)
- Jing Liu
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA
| | - Zsu-Zsu Chen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA
| | - Jagvi Patel
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA
| | - Aarti Asnani
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA.
| |
Collapse
|
5
|
de Boer RA, Aboumsallem JP, Bracun V, Leedy D, Cheng R, Patel S, Rayan D, Zaharova S, Rymer J, Kwan JM, Levenson J, Ronco C, Thavendiranathan P, Brown SA. A new classification of cardio-oncology syndromes. CARDIO-ONCOLOGY 2021; 7:24. [PMID: 34154667 PMCID: PMC8218489 DOI: 10.1186/s40959-021-00110-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/30/2021] [Indexed: 12/21/2022]
Abstract
Increasing evidence suggests a multifaceted relationship exists between cancer and cardiovascular disease (CVD). Here, we introduce a 5-tier classification system to categorize cardio-oncology syndromes (COS) that represent the aspects of the relationship between cancer and CVD. COS Type I is characterized by mechanisms whereby the abrupt onset or progression of cancer can lead to cardiovascular dysfunction. COS Type II includes the mechanisms by which cancer therapies can result in acute or chronic CVD. COS Type III is characterized by the pro-oncogenic environment created by the release of cardiokines and high oxidative stress in patients with cardiovascular dysfunction. COS Type IV is comprised of CVD therapies and diagnostic procedures which have been associated with promoting or unmasking cancer. COS Type V is characterized by factors causing systemic and genetic predisposition to both CVD and cancer. The development of this framework may allow for an increased facilitation of cancer care while optimizing cardiovascular health through focused treatment targeting the COS type.
Collapse
Affiliation(s)
- Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Valentina Bracun
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Douglas Leedy
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Richard Cheng
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Sahishnu Patel
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - David Rayan
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Svetlana Zaharova
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | | | - Jennifer M Kwan
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Joshua Levenson
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Claudio Ronco
- Department of Medicine, University of Padova, Padova, Italy.,International Renal Research Institute of Vicenza, Vicenza, Italy.,Department of Nephrology, San Bortolo Hospital, Vicenza, Italy
| | | | - Sherry-Ann Brown
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
6
|
Kamaraju S, Mohan M, Zaharova S, Wallace B, McGraw J, Lokken J, Tierney J, Weil E, Fatunde O, Brown SA. Interactions between cardiology and oncology drugs in precision cardio-oncology. Clin Sci (Lond) 2021; 135:1333-1351. [PMID: 34076246 PMCID: PMC8984624 DOI: 10.1042/cs20200309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/26/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022]
Abstract
Recent advances in treatment have transformed the management of cancer. Despite these advances, cardiovascular disease remains a leading cause of death in cancer survivors. Cardio-oncology has recently evolved as a subspecialty to prevent, diagnose, and manage cardiovascular side effects of antineoplastic therapy. An emphasis on optimal management of comorbidities and close attention to drug interactions are important in cardio-oncologic care. With interdisciplinary collaboration among oncologists, cardiologists, and pharmacists, there is potential to prevent and reduce drug-related toxicities of treatments. The cytochrome P450 (CYP450) family of enzymes and the P-glycoprotein (P-g) transporter play a crucial role in drug metabolism and drug resistance. Here we discuss the role of CYP450 and P-g in drug interactions in the field of cardio-oncology, provide an overview of the cardiotoxicity of a spectrum of cancer agents, highlight the role of precision medicine, and encourage a multidisciplinary treatment approach for patients with cancer.
Collapse
Affiliation(s)
- Sailaja Kamaraju
- Division of Hematology and Oncology, Department of
Medicine, Medical College of Wisconsin, WI, U.S.A
| | - Meera Mohan
- Division of Hematology and Oncology, Department of
Medicine, Medical College of Wisconsin, WI, U.S.A
| | - Svetlana Zaharova
- Cardio-Oncology Program, Division of Cardiovascular
Medicine, Medical College of Wisconsin, Milwaukee, WI, U.S.A
| | | | - Joseph McGraw
- Department of Pharmacy, Concordia University, Milwaukee,
WI, U.S.A
| | - James Lokken
- Department of Pharmacy, Concordia University, Milwaukee,
WI, U.S.A
| | - John Tierney
- School of Pharmacy, Medical College of Wisconsin, WI,
U.S.A
| | - Elizabeth Weil
- Department of Pharmacy, Medical College of Wisconsin, WI,
U.S.A
| | - Olubadewa Fatunde
- Division of Cardiology, Department of Medicine, Mayo Clinic
Arizona, Scottsdale, AZ, U.S.A
| | - Sherry-Ann Brown
- Cardio-Oncology Program, Division of Cardiovascular
Medicine, Medical College of Wisconsin, Milwaukee, WI, U.S.A
| |
Collapse
|
7
|
Modeling Precision Cardio-Oncology: Using Human-Induced Pluripotent Stem Cells for Risk Stratification and Prevention. Curr Oncol Rep 2021; 23:77. [PMID: 33937943 PMCID: PMC8088904 DOI: 10.1007/s11912-021-01066-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 11/12/2022]
Abstract
Purpose of Review Cardiovascular toxicity is a leading cause of mortality among cancer survivors and has become increasingly prevalent due to improved cancer survival rates. In this review, we synthesize evidence illustrating how common cancer therapeutic agents, such as anthracyclines, human epidermal growth factors receptors (HER2) monoclonal antibodies, and tyrosine kinase inhibitors (TKIs), have been evaluated in cardiomyocytes (CMs) derived from human-induced pluripotent stem cells (hiPSCs) to understand the underlying mechanisms of cardiovascular toxicity. We place this in the context of precision cardio-oncology, an emerging concept for personalizing the prevention and management of cardiovascular toxicities from cancer therapies, accounting for each individual patient’s unique factors. We outline steps that will need to be addressed by multidisciplinary teams of cardiologists and oncologists in partnership with regulators to implement future applications of hiPSCs in precision cardio-oncology. Recent Findings Current prevention of cardiovascular toxicity involves routine screenings and management of modifiable risk factors for cancer patients, as well as the initiation of cardioprotective medications. Despite recent advancements in precision cardio-oncology, knowledge gaps remain and limit our ability to appropriately predict with precision which patients will develop cardiovascular toxicity. Investigations using patient-specific CMs facilitate pharmacological discovery, mechanistic toxicity studies, and the identification of cardioprotective pathways. Studies with hiPSCs demonstrate that patients with comorbidities have more frequent adverse responses, compared to their counterparts without cardiac disease. Further studies utilizing hiPSC modeling should be considered, to evaluate the impact and mitigation of known cardiovascular risk factors, including blood pressure, body mass index (BMI), smoking status, diabetes, and physical activity in their role in cardiovascular toxicity after cancer therapy. Future real-world applications will depend on understanding the current use of hiPSC modeling in order for oncologists and cardiologists together to inform their potential to improve our clinical collaborative practice in cardio-oncology. Summary When applying such in vitro characterization, it is hypothesized that a safety score can be assigned to each individual to determine who has a greater probability of developing cardiovascular toxicity. Using hiPSCs to create personalized models and ultimately evaluate the cardiovascular toxicity of individuals’ treatments may one day lead to more patient-specific treatment plans in precision cardio-oncology while reducing cardiovascular disease (CVD) morbidity and mortality.
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW BRAF/MEK inhibitor has changed the treatment landscape in patients with advanced and metastatic melanoma with prolonged overall survival and progression-free survival. Since three treatment combinations exist with similar efficacy therapy decisions are often made based on the side effect profile. Additionally, on-target side effects or class effects have to be properly managed to ensure treatment adherence. RECENT FINDINGS Sequential treatment with BRAF/MEK inhibition and immunotherapy might increase toxicity with a sepsis-like syndrome and triple therapy with concomitant BRAF/MEK inhibition and anti-PD1/PD-L1 antibody therapy induces severe side effects in the vast majority of patients. SUMMARY Toxicity of combination therapy with BRAF/MEK inhibitors is generally manageable, reversible and infrequently associated with treatment discontinuation. In case of persisting off-target effects the change to another combination therapy can resolve side effects.
Collapse
Affiliation(s)
- Alvaro Moreira
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai
- The Kimberly and Eric J. Waldman Department of Dermatology at Mount Sinai, New York, NY, USA
| | - Céleste Lebbé
- Université de Paris, AP-HP Dermatology, INSERM U976, Saint Louis Hospital, Paris, France
| | - Lucie Heinzerling
- Department of Dermatology, Universitätsklinikum München (LMU), Munich, Germany
- Department of Dermatology, Universitätsklinikum Erlangen, Germany and Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
| |
Collapse
|
9
|
Iacopo F, Branch M, Cardinale D, Middeldorp M, Sanders P, Cohen JB, Achirica MC, Jaiswal S, Brown SA. Preventive Cardio-Oncology: Cardiovascular Disease Prevention in Cancer Patients and Survivors. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2021. [DOI: 10.1007/s11936-020-00883-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
10
|
Brown SA, Zaharova S, Mason P, Thompson J, Thapa B, Ishizawar D, Wilkes E, Ahmed G, Rubenstein J, Sanchez J, Joyce D, Kalyanaraman B, Widlansky M. Pandemic Perspective: Commonalities Between COVID-19 and Cardio-Oncology. Front Cardiovasc Med 2020; 7:568720. [PMID: 33344513 PMCID: PMC7746643 DOI: 10.3389/fcvm.2020.568720] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022] Open
Abstract
Overlapping commonalities between coronavirus disease of 2019 (COVID-19) and cardio-oncology regarding cardiovascular toxicities (CVT), pathophysiology, and pharmacology are special topics emerging during the pandemic. In this perspective, we consider an array of CVT common to both COVID-19 and cardio-oncology, including cardiomyopathy, ischemia, conduction abnormalities, myopericarditis, and right ventricular (RV) failure. We also emphasize the higher risk of severe COVID-19 illness in patients with cardiovascular disease (CVD) or its risk factors or cancer. We explore commonalities in the underlying pathophysiology observed in COVID-19 and cardio-oncology, including inflammation, cytokine release, the renin-angiotensin-aldosterone-system, coagulopathy, microthrombosis, and endothelial dysfunction. In addition, we examine common pharmacologic management strategies that have been elucidated for CVT from COVID-19 and various cancer therapies. The use of corticosteroids, as well as antibodies and inhibitors of various molecules mediating inflammation and cytokine release syndrome, are discussed. The impact of angiotensin converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) is also addressed, since these drugs are used in cardio-oncology and have received considerable attention during the COVID-19 pandemic, since the culprit virus enters human cells via the angiotensin converting enzyme 2 (ACE2) receptor. There are therefore several areas of overlap, similarity, and interaction in the toxicity, pathophysiology, and pharmacology profiles in COVID-19 and cardio-oncology syndromes. Learning more about either will likely provide some level of insight into both. We discuss each of these topics in this viewpoint, as well as what we foresee as evolving future directions to consider in cardio-oncology during the pandemic and beyond. Finally, we highlight commonalities in health disparities in COVID-19 and cardio-oncology and encourage continued development and implementation of innovative solutions to improve equity in health and healing.
Collapse
Affiliation(s)
- Sherry-Ann Brown
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Svetlana Zaharova
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Peter Mason
- Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jonathan Thompson
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bicky Thapa
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - David Ishizawar
- Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Erin Wilkes
- Department of Pharmacy, Froedtert Health and Medical College of Wisconsin, Milwaukee, WI, United States
| | - Gulrayz Ahmed
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jason Rubenstein
- Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Joyce Sanchez
- Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - David Joyce
- Division of Cardiothoracic Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Michael Widlansky
- Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
11
|
Abu Rmilah AA, Lin G, Begna KH, Friedman PA, Herrmann J. Risk of QTc prolongation among cancer patients treated with tyrosine kinase inhibitors. Int J Cancer 2020; 147:3160-3167. [PMID: 32449208 PMCID: PMC8772338 DOI: 10.1002/ijc.33119] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/31/2022]
Abstract
QTc interval prolongation can lead to life-threatening complications such as Torsade de Pointes (TdP), ventricular tachycardia (VT) and sudden cardiac death (SCD). It can occur with tyrosine kinase inhibitors (TKIs) but comparative real-world analyses on the incidence and complication rates are scarce. We retrospectively reviewed all cancer patients treated with TKI therapy at Mayo Clinic between January 2005 and December 2018 and had at least two ECGs (before and after TKI). For each TKI type, we determined the administration rate and incidence of QTc prolongation. QTc prolongation was defined as a corrected QT interval (by Fridericia formula) ≥450 ms in men and ≥470 ms in women. A total of 618 cancer patients were included with 902 TKI administrations, of which 654 (72.5%) were accounted for by pazopanib, sunitinib, imatinib, nilotinib and dasatinib. QTc prolongation (any grade) was reported in 28.8%, most commonly with nilotinib (38.7%) and dasatinib (41.7%). A QTc interval ≥500 ms and a QTc increase ≥60 ms was documented in 46 and 63 administrations, respectively. Life-threatening toxicity was seen in 14 cases (5.4% of QTc prolongation cases) including VT in 9, SCD in 3 and TdP in two administrations. The response to QTc prolongation was: discontinuation in 68%, dose reduction in 13.5%, temporary hold in 8.1% and no action in 10.4%. In conclusion, QTc prolongation with TKI therapy is very common (∼1/3 of cases) and in 5% (1.7% overall) associated with life-threatening complications. These data support recommendations for careful ECG monitoring in cancer patients undergoing TKI therapy.
Collapse
Affiliation(s)
- Anan A. Abu Rmilah
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Grace Lin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kebede H. Begna
- Department of Internal Medicine, Division of Hematology, Mayo Clinic, Rochester, Minnesota
| | - Paul A. Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
12
|
Abstract
OPINION STATEMENT As the world becomes more connected through online and offline social networking, there has been much discussion of how the rapid rise of social media could be used in ways that can be productive and instructive in various healthcare specialties, such as Cardiology and its subspecialty areas. In this review, the role of social media in the field of Cardio-Oncology is discussed. With an estimated 17 million cancer survivors in the USA in 2019 and 22 million estimated by 2030, more education and awareness are needed. Networking and collaboration are also needed to meet the needs of our patients and healthcare professionals in this emerging field bridging two disciplines. Cardiovascular disease is second only to recurrence of the primary cancer or diagnosis with a secondary malignancy, as a leading cause of death in cancer survivors. A majority of these survivors are anticipated to be on social media seeking information, support, and ideas for optimizing health. Healthcare professionals in Cardio-Oncology are also online for networking, education, scholarship, career development, and advocacy in this field. Here, we describe the utilization and potential impact of social media in Cardio-Oncology, with inclusion of various hashtags frequently used in the Cardio-Oncology Twitter community.
Collapse
|
13
|
Abstract
Remarkable progress has been made in the development of new therapies for cancer, dramatically changing the landscape of treatment approaches for several malignancies and continuing to increase patient survival. Accordingly, adverse effects of cancer therapies that interfere with the continuation of best-possible care, induce life-threatening risks or lead to long-term morbidity are gaining increasing importance. Cardiovascular toxic effects of cancer therapeutics and radiation therapy are the epitome of such concerns, and proper knowledge, interpretation and management are needed and have to be placed within the context of the overall care of individual patients with cancer. Furthermore, the cardiotoxicity spectrum has broadened to include myocarditis with immune checkpoint inhibitors and cardiac dysfunction in the setting of cytokine release syndrome with chimeric antigen receptor T cell therapy. An increase in the incidence of arrhythmias related to inflammation such as atrial fibrillation can also be expected, in addition to the broadening set of cancer therapeutics that can induce prolongation of the corrected QT interval. Therefore, cardiologists of today have to be familiar not only with the cardiotoxicity associated with traditional cancer therapies, such as anthracycline, trastuzumab or radiation therapy, but even more so with an ever-increasing repertoire of therapeutics. This Review provides this information, summarizing the latest developments at the juncture of cardiology, oncology and haematology.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
14
|
Brown SA, Ray JC, Herrmann J. Precision Cardio-Oncology: a Systems-Based Perspective on Cardiotoxicity of Tyrosine Kinase Inhibitors and Immune Checkpoint Inhibitors. J Cardiovasc Transl Res 2020; 13:402-416. [PMID: 32253744 PMCID: PMC8855704 DOI: 10.1007/s12265-020-09992-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 03/18/2020] [Indexed: 02/07/2023]
Abstract
Cancer therapies have been evolving from conventional chemotherapeutics to targeted agents. This has fulfilled the hope of greater efficacy but unfortunately not of greater safety. In fact, a broad spectrum of toxicities can be seen with targeted therapies, including cardiovascular toxicities. Among these, cardiomyopathy and heart failure have received greatest attention, given their profound implications for continuation of cancer therapies and cardiovascular morbidity and mortality. Prediction of risk has always posed a challenge and even more so with the newer targeted agents. The merits of accurate risk prediction, however, are very evident, e.g. facilitating treatment decisions even before the first dose is given. This is important for agents with a long half-life and high potential to induced life-threatening cardiac complications, such as myocarditis with immune checkpoint inhibitors. An opportunity to address these needs in the field of cardio-oncology is provided by the expanding repertoire of "-omics" and other tools in precision medicine and their integration in a systems biology approach. This may allow for new insights into patho-mechanisms and the creation of more precise and cost-effective risk prediction tools with the ultimate goals of improved therapy decisions and prevention of cardiovascular complications. Herein, we explore this topic as a future approach to translating the complexity of cardio-oncology to the reality of patient care.
Collapse
Affiliation(s)
- Sherry-Ann Brown
- Department of Cardiovascular Diseases, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Jordan C Ray
- Department of Cardiovascular Diseases, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
15
|
Fatunde OA, Brown SA. The Role of CYP450 Drug Metabolism in Precision Cardio-Oncology. Int J Mol Sci 2020; 21:E604. [PMID: 31963461 PMCID: PMC7014347 DOI: 10.3390/ijms21020604] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
As many novel cancer therapies continue to emerge, the field of Cardio-Oncology (or onco-cardiology) has become crucial to prevent, monitor and treat cancer therapy-related cardiovascular toxicity. Furthermore, given the narrow therapeutic window of most cancer therapies, drug-drug interactions are prevalent in the cancer population. Consequently, there is an increased risk of affecting drug efficacy or predisposing individual patients to adverse side effects. Here we review the role of cytochrome P450 (CYP450) enzymes in the field of Cardio-Oncology. We highlight the importance of cardiac medications in preventive Cardio-Oncology for high-risk patients or in the management of cardiotoxicities during or following cancer treatment. Common interactions between Oncology and Cardiology drugs are catalogued, emphasizing the impact of differential metabolism of each substrate drug on unpredictable drug bioavailability and consequent inter-individual variability in treatment response or development of cardiovascular toxicity. This inter-individual variability in bioavailability and subsequent response can be further enhanced by genomic variants in CYP450, or by modifications of CYP450 gene, RNA or protein expression or function in various 'omics' related to precision medicine. Thus, we advocate for an individualized approach to each patient by a multidisciplinary team with clinical pharmacists evaluating a treatment plan tailored to a practice of precision Cardio-Oncology. This review may increase awareness of these key concepts in the rapidly evolving field of Cardio-Oncology.
Collapse
Affiliation(s)
- Olubadewa A. Fatunde
- Department of Medicine, University of Texas Health Science Center at Tyler–CHRISTUS Good Shepherd Medical Center, Longview, TX 75601, USA
| | - Sherry-Ann Brown
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
16
|
Ma W, Liu M, Liang F, Zhao L, Gao C, Jiang X, Zhang X, Zhan H, Hu H, Zhao Z. Cardiotoxicity of sorafenib is mediated through elevation of ROS level and CaMKII activity and dysregulation of calcium homoeostasis. Basic Clin Pharmacol Toxicol 2019; 126:166-180. [DOI: 10.1111/bcpt.13318] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Wenzhuo Ma
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Mei Liu
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Fanfan Liang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Lili Zhao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Chenying Gao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Xixi Jiang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Xin Zhang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Heqin Zhan
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
- Department of Pharmacology College of Pharmacy Xinxiang Medical University Xinxiang Henan 453003 China
| | - Hao Hu
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Zhenghang Zhao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| |
Collapse
|
17
|
Lai Y, Zhao Z, Zeng T, Liang X, Chen D, Duan X, Zeng G, Wu W. Crosstalk between VEGFR and other receptor tyrosine kinases for TKI therapy of metastatic renal cell carcinoma. Cancer Cell Int 2018. [PMID: 29527128 PMCID: PMC5838927 DOI: 10.1186/s12935-018-0530-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cell carcinoma (RCC), and is frequently accompanied by the genetic features of von Hippel–Lindau (VHL) loss. VHL loss increases the expression of hypoxia-inducible factors (HIFs) and their targets, including epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF). The primary treatment for metastatic RCC (mRCC) is molecular-targeted therapy, especially anti-angiogenic therapy. VEGF monoclonal antibodies and VEGF receptor (VEGFR) tyrosine kinase inhibitors (TKIs) are the main drugs used in anti-angiogenic therapy. However, crosstalk between VEGFR and other tyrosine kinase or downstream pathways produce resistance to TKI treatment, and the multi-target inhibitors, HIF inhibitors or combination strategies are promising strategies for mRCC. HIFs are upstream of the crosstalk between the growth factors, and these factors may regulate the expression of VEGR, EGF, PDGF and other growth factors. The frequent VHL loss in ccRCC increases HIF expression, and HIFs may be an ideal candidate to overcome the TKI resistance. The combination of HIF inhibitors and immune checkpoint inhibitors is also anticipated. Various clinical trials of programmed cell death protein 1 inhibitors are planned. The present study reviews the effects of current and potential TKIs on mRCC, with a focus on VEGF/VEGFR and other targets for mRCC therapy.
Collapse
Affiliation(s)
- Yongchang Lai
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Zhijian Zhao
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Tao Zeng
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Xiongfa Liang
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Dong Chen
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Xiaolu Duan
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Guohua Zeng
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Wenqi Wu
- Department of Urology, Minimally Invasive Surgery Center, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| |
Collapse
|
18
|
Mayekar MK, Bivona TG. Current Landscape of Targeted Therapy in Lung Cancer. Clin Pharmacol Ther 2017; 102:757-764. [PMID: 28786099 DOI: 10.1002/cpt.810] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022]
Abstract
Lung cancer is the leading cause of cancer mortality worldwide. Comprehensive genomic profiling of lung cancers revealed their genetic heterogeneity and complexity and identified numerous targetable oncogenic driver alterations. These molecular profiling efforts have made it possible to exploit the potential of molecularly targeted therapies. Selection of patients for targeted therapies is becoming biomarker-driven, where the oncogenic drivers in patient tumors are first identified, and subsequently patients bearing drug-sensitizing genetic aberrations are matched to the appropriate targeted therapy. Success of this design of clinical trials and practice was first demonstrated in EGFR inhibitor trials in lung cancer and has since been incorporated into subsequent targeted therapy trials including ALK-, ROS1-, and BRAF V600E-targeted therapies. In this review we discuss the current landscape of clinically approved and other promising molecularly targeted approaches for the treatment of lung cancers, the challenges with these approaches, and the strategies that could be deployed to overcome these challenges.
Collapse
Affiliation(s)
- Manasi K Mayekar
- Department of Medicine, University of California, San Francisco, San Francisco, USA.,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, USA.,Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, USA
| |
Collapse
|
19
|
Aghel N, Delgado DH, Lipton JH. Cardiovascular toxicities of BCR-ABL tyrosine kinase inhibitors in chronic myeloid leukemia: preventive strategies and cardiovascular surveillance. Vasc Health Risk Manag 2017; 13:293-303. [PMID: 28831263 PMCID: PMC5552150 DOI: 10.2147/vhrm.s108874] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment and outcomes of chronic myeloid leukemia (CML). Despite their significant impact on the management of CML, there is growing evidence that TKIs may cause cardiovascular and/or metabolic complications. In this review, we present the current evidence regarding the cardiovascular safety profiles of BCR-ABL TKIs. Methodological challenges of studies that reported the cardiovascular safety of TKIs are discussed. We also propose management strategies for cardiovascular surveillance and risk factor modification during treatment with these agents.
Collapse
Affiliation(s)
- Nazanin Aghel
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network,University of Toronto
| | - Diego Hernan Delgado
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network,University of Toronto
| | - Jeffrey Howard Lipton
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|