1
|
Senechal I, Andres MS, Tong J, Ramalingam S, Nazir MS, Rosen SD, Young K, Idaikkadar P, Larkin J, Lyon AR. Risk Stratification, Screening and Treatment of BRAF/MEK Inhibitors-Associated Cardiotoxicity. Curr Oncol Rep 2024:10.1007/s11912-024-01599-2. [PMID: 39316222 DOI: 10.1007/s11912-024-01599-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE OF REVIEW In this review article we describe the cardiovascular adverse events associated with BRAF and MEK inhibitors as well as their pathophysiologic mechanisms and provide up to date guidance for risk stratified surveillance of patients on treatment and the optimal management of emergent cardiotoxicities. RECENT FINDINGS Combination BRAF/MEK inhibition has become an established standard treatment option for patients with a wide variety of BRAF mutant haematological and solid organ cancers, its use is most commonly associated with stage three and metastatic melanoma. The introduction of these targeted drugs has significantly improved the prognosis of previously treatment resistant cancers. It is increasingly recognised that these drugs have a number of cardiovascular toxicities including left ventricular systolic dysfunction, hypertension and QTc interval prolongation. Whilst cardiotoxicity is largely reversible and manageable with medical therapy, it does limit the effective use of these highly active agents.
Collapse
Affiliation(s)
- Isabelle Senechal
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK.
- Centre Hospitalier Universitaire de Québec, Québec City, Québec, Canada.
| | - Maria Sol Andres
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Jieli Tong
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Muhummad Sohaib Nazir
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Stuart D Rosen
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kate Young
- Royal Marsden Hospital Foundation Trust, London, UK
| | | | - James Larkin
- Royal Marsden Hospital Foundation Trust, London, UK
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
2
|
Elshazly AM, Xu J, Melhem N, Abdulnaby A, Elzahed AA, Saleh T, Gewirtz DA. Is Autophagy Targeting a Valid Adjuvant Strategy in Conjunction with Tyrosine Kinase Inhibitors? Cancers (Basel) 2024; 16:2989. [PMID: 39272847 PMCID: PMC11394573 DOI: 10.3390/cancers16172989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) represent a relatively large class of small-molecule inhibitors that compete with ATP for the catalytic binding site of tyrosine kinase proteins. While TKIs have demonstrated effectiveness in the treatment of multiple malignancies, including chronic myelogenous leukemia, gastrointestinal tumors, non-small cell lung cancers, and HER2-overexpressing breast cancers, as is almost always the case with anti-neoplastic agents, the development of resistance often imposes a limit on drug efficacy. One common survival response utilized by tumor cells to ensure their survival in response to different stressors, including anti-neoplastic drugs, is that of autophagy. The autophagic machinery in response to TKIs in multiple tumor models has largely been shown to be cytoprotective in nature, although there are a number of cases where autophagy has demonstrated a cytotoxic function. In this review, we provide an overview of the literature examining the role that autophagy plays in response to TKIs in different preclinical tumor model systems in an effort to determine whether autophagy suppression or modulation could be an effective adjuvant strategy to increase efficiency and/or overcome resistance to TKIs.
Collapse
Affiliation(s)
- Ahmed M Elshazly
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Jingwen Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Nebras Melhem
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | - Alsayed Abdulnaby
- Department of Pharmacognosy, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Aya A Elzahed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, Hashemite University, Zarqa 13133, Jordan
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA
| |
Collapse
|
3
|
Tetterton-Kellner J, Jensen BC, Nguyen J. Navigating cancer therapy induced cardiotoxicity: From pathophysiology to treatment innovations. Adv Drug Deliv Rev 2024; 211:115361. [PMID: 38901637 DOI: 10.1016/j.addr.2024.115361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
Every year, more than a million people in the United States undergo chemotherapy or radiation therapy for cancer, as estimated by the CDC. While chemotherapy has been an instrumental tool for treating cancer, it also causes severe adverse effects. The more commonly acknowledged adverse effects include hair loss, fatigue, and nausea, but a more severe and longer lasting side effect is cardiotoxicity. Cardiotoxicity, or heart damage, is a common complication of cancer treatments. It can range from mild to severe, and it can affect some patients temporarily or others permanently, even after they are cured of cancer. Dexrazoxane is the only FDA-approved drug for treating anthracycline induced cardiotoxicity, but it also has drawbacks and adverse effects. There is no other type of chemotherapy induced cardiotoxicity that has an approved treatment option. In this review, we discuss the pathophysiology of chemotherapeutic-induced cardiotoxicity, methods and guidelines of diagnosis, methods of treatment and mitigation, and current drug delivery approaches in therapeutic development.
Collapse
Affiliation(s)
- Jessica Tetterton-Kellner
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian C Jensen
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Cardiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
4
|
Grogan L, Shapiro P. Progress in the development of ERK1/2 inhibitors for treating cancer and other diseases. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 100:181-207. [PMID: 39034052 DOI: 10.1016/bs.apha.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
The extracellular signal-regulated kinases-1 and 2 (ERK1/2) are ubiquitous regulators of many cellular functions, including proliferation, differentiation, migration, and cell death. ERK1/2 regulate cell functions by phosphorylating a diverse collection of protein substrates consisting of other kinases, transcription factors, structural proteins, and other regulatory proteins. ERK1/2 regulation of cell functions is tightly regulated through the balance between activating phosphorylation by upstream kinases and inactivating dephosphorylation by phosphatases. Disruption of homeostatic ERK1/2 regulation caused by elevated extracellular signals or mutations in upstream regulatory proteins leads to the constitutive activation of ERK1/2 signaling and uncontrolled cell proliferation observed in many types of cancer. Many inhibitors of upstream kinase regulators of ERK1/2 have been developed and are part of targeted therapeutic options to treat a variety of cancers. However, the efficacy of these drugs in providing sustained patient responses is limited by the development of acquired resistance often involving re-activation of ERK1/2. As such, recent drug discovery efforts have focused on the direct targeting of ERK1/2. Several ATP competitive ERK1/2 inhibitors have been identified and are being tested in cancer clinical trials. One drug, Ulixertinib (BVD-523), has received FDA approval for use in the Expanded Access Program for patients with no other therapeutic options. This review provides an update on ERK1/2 inhibitors in clinical trials, their successes and limitations, and new academic drug discovery efforts to modulate ERK1/2 signaling for treating cancer and other diseases.
Collapse
Affiliation(s)
- Lena Grogan
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States.
| |
Collapse
|
5
|
Abboud K, Umoru G, Trachtenberg B, Ajewole V. Real-world data of cardio-oncologic interventions for cardiovascular adverse events with oral oncolytics. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:22. [PMID: 38594785 PMCID: PMC11003064 DOI: 10.1186/s40959-024-00221-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Oral cancer therapy-related cardiovascular (CV) toxicity has a wide variety of presentations including arrhythmia, cardiomyopathy, and myocardial infarction, but clinical evidence related to its management is limited. The purpose of this IRB-approved, single-center, retrospective, cohort study was to characterize cardio-oncologic interventions for CV adverse events related to oral oncolytics. METHODS The cohort included 67 patients who were admitted to a multi-hospital health system between June 1, 2016 and July 31, 2021, had at least one medical record order of oral oncolytics considered to have cardiotoxic potential, and had an ICD10 code for a cardiotoxic event added to their electronic medical records after initiation of oral oncolytics. RESULTS The majority (97%) had pre-existing cardiovascular disease (CVD) or a CV risk factor. The three most common classes of oral oncolytics were aromatase inhibitors (36%), BCR-ABL inhibitors (16%), and VEGFR inhibitors (13%). New-onset or worsening heart failure (HF) (n = 31), which occurred after a median of 148 days (Interquartile range (IQR) 43-476 days) was the most common cardiotoxic event. The most frequent interventions were pharmacological treatment of the CV adverse event (n = 44) and treatment interruption (n = 18), but guideline-directed medication therapy for HF could be further optimized. CONCLUSION Pre-existing CVD or CV risk factors predispose oncology patients to CV adverse events. Real-world practice reveals that CV adverse events require temporary interruption of treatment and initiation of pharmacologic treatment. A multidisciplinary, patient-centered approach that includes discussion of risks/benefits of treatment continuation, and initiation of guideline-directed treatment is recommended until high-quality, drug-specific data for monitoring and treatment become available.
Collapse
Affiliation(s)
- Karen Abboud
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, USA
| | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, USA
| | - Barry Trachtenberg
- Heart Failure and Transplantation Cardiology, Houston Methodist Hospital, Houston, TX, USA
| | - Veronica Ajewole
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, USA.
- Texas Southern University College of Pharmacy & Health Sciences, Houston, TX, USA.
| |
Collapse
|
6
|
Kreidieh F, McQuade J. Novel insights into cardiovascular toxicity of cancer targeted and immune therapies: Beyond ischemia with non-obstructive coronary arteries (INOCA). AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 40:100374. [PMID: 38510501 PMCID: PMC10946000 DOI: 10.1016/j.ahjo.2024.100374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
Novel immune and targeted therapies approved over the past 2 decades have resulted in dramatic improvements in cancer-specific outcomes for many cancer patients. However, many of these agents can induce cardiovascular toxicity in a subset of patients. The field of cardio-oncology was established based on observations that anti-neoplastic chemotherapies and mantle radiation can lead to premature cardiomyopathy in cancer survivors. While conventional chemotherapy, targeted therapy, and immune therapies can all result in cardiovascular adverse events, the mechanisms, timing, and incidence of these events are inherently different. Many of these effects converge upon the coronary microvasculature to involve, through endocardial endothelial cells, a more direct effect through close proximity to cardiomyocyte with cellular communication and signaling pathways. In this review, we will provide an overview of emerging paradigms in the field of Cardio-Oncology, particularly the role of the coronary microvasculature in mediating cardiovascular toxicity of important cancer targeted and immune therapies. As the number of cancer patients treated with novel immune and targeted therapies grows exponentially and subsequently the number of long-term cancer survivors dramatically increases, it is critical that cardiologists and cardiology researchers recognize the unique potential cardiovascular toxicities of these agents.
Collapse
Affiliation(s)
- Firas Kreidieh
- Instructor of Clinical Medicine- Division of Hematology-Oncology; Associate Director- Internal Medicine Residency Program, American University of Beirut, Beirut, Lebanon
| | - Jennifer McQuade
- Associate Professor and Physician Scientist in Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
7
|
Ureña E, Xu B, Regan JC, Atilano ML, Minkley LJ, Filer D, Lu YX, Bolukbasi E, Khericha M, Alic N, Partridge L. Trametinib ameliorates aging-associated gut pathology in Drosophila females by reducing Pol III activity in intestinal stem cells. Proc Natl Acad Sci U S A 2024; 121:e2311313121. [PMID: 38241436 PMCID: PMC10823232 DOI: 10.1073/pnas.2311313121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/17/2023] [Indexed: 01/21/2024] Open
Abstract
Pharmacological therapies are promising interventions to slow down aging and reduce multimorbidity in the elderly. Studies in animal models are the first step toward translation of candidate molecules into human therapies, as they aim to elucidate the molecular pathways, cellular mechanisms, and tissue pathologies involved in the anti-aging effects. Trametinib, an allosteric inhibitor of MEK within the Ras/MAPK (Ras/Mitogen-Activated Protein Kinase) pathway and currently used as an anti-cancer treatment, emerged as a geroprotector candidate because it extended lifespan in the fruit fly Drosophila melanogaster. Here, we confirm that trametinib consistently and robustly extends female lifespan, and reduces intestinal stem cell (ISC) proliferation, tumor formation, tissue dysplasia, and barrier disruption in guts in aged flies. In contrast, pro-longevity effects of trametinib are weak and inconsistent in males, and it does not influence gut homeostasis. Inhibition of the Ras/MAPK pathway specifically in ISCs is sufficient to partially recapitulate the effects of trametinib. Moreover, in ISCs, trametinib decreases the activity of the RNA polymerase III (Pol III), a conserved enzyme synthesizing transfer RNAs and other short, non-coding RNAs, and whose inhibition also extends lifespan and reduces gut pathology. Finally, we show that the pro-longevity effect of trametinib in ISCs is partially mediated by Maf1, a repressor of Pol III, suggesting a life-limiting Ras/MAPK-Maf1-Pol III axis in these cells. The mechanism of action described in this work paves the way for further studies on the anti-aging effects of trametinib in mammals and shows its potential for clinical application in humans.
Collapse
Affiliation(s)
- Enric Ureña
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Bowen Xu
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Jennifer C. Regan
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Magda L. Atilano
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Lucy J. Minkley
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Danny Filer
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Yu-Xuan Lu
- Max Planck Institute for Biology of Ageing, CologneD-50931, Germany
| | - Ekin Bolukbasi
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Mobina Khericha
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Nazif Alic
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Linda Partridge
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
- Max Planck Institute for Biology of Ageing, CologneD-50931, Germany
| |
Collapse
|
8
|
Pavlovic D, Niciforovic D, Papic D, Milojevic K, Markovic M. CDK4/6 inhibitors: basics, pros, and major cons in breast cancer treatment with specific regard to cardiotoxicity - a narrative review. Ther Adv Med Oncol 2023; 15:17588359231205848. [PMID: 37841752 PMCID: PMC10571689 DOI: 10.1177/17588359231205848] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Breast cancer is characterized by the uncontrolled proliferation of breast cells, with a high incidence reported in 2020 to have affected over 2 million women. In recent years, the conventional methods of treating breast cancer have involved radiotherapy and chemotherapy. However, the emergence of CDK4/6 inhibitors has shown potential as a promising cancer therapy. Cyclin-dependent kinases (CDK) inhibitors are a class of molecules that impede the formation of an active kinase complex, thereby hindering its activity and consequently halting the progression of the cell cycle. It was discovered that they have a significant impact on impeding the progression of the cancer. This is evident with the Food and Drug Administration's approval of drugs such as palbociclib, ribociclib, and abemaciclib for hormone receptor-positive metastatic breast cancer in combination with specific endocrine therapies. In spite of enormous success in breast cancer treatment, certain obstacles have emerged, such as therapy resistance, side effects, and most of all, cardiotoxicity. Some of these drawbacks have been successfully overcome by dosage reduction, different combinations of the drugs, and the assessment of each patient's condition and suitability prior to treatment. Yet other drawbacks still require tenacious research, especially certain cases of cardiotoxicities. This article delves into the biological mechanisms of CDK4/6 in the cell cycle and cancer, as well as the clinical advantages and most common adverse events (AEs) associated with CDK4/6 inhibitors. The primary objective of this review is to provide a comprehensive analysis of cardiotoxic AEs and elucidate the underlying pathophysiological mechanisms responsible for the cardiotoxicity of CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Dragica Pavlovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, Kragujevac 34000, Serbia
| | - Danijela Niciforovic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Dragana Papic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Katarina Milojevic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marina Markovic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
9
|
Gross AM, Dombi E, Wolters PL, Baldwin A, Dufek A, Herrera K, Martin S, Derdak J, Heisey KS, Whitcomb PM, Steinberg SM, Venzon DJ, Fisher MJ, Kim A, Bornhorst M, Weiss BD, Blakeley JO, Smith MA, Widemann BC. Long-term safety and efficacy of selumetinib in children with neurofibromatosis type 1 on a phase 1/2 trial for inoperable plexiform neurofibromas. Neuro Oncol 2023; 25:1883-1894. [PMID: 37115514 PMCID: PMC10547508 DOI: 10.1093/neuonc/noad086] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Selumetinib shrank inoperable symptomatic plexiform neurofibromas (PN) in children with neurofibromatosis type 1 (NF1) and provided clinical benefit for many in our previously published phase 1/2 clinical trials (SPRINT, NCT01362803). At the data cutoff (DCO) of the prior publications, 65% of participants were still receiving treatment. This report presents up to 5 years of additional safety and efficacy data from these studies. METHODS This manuscript includes data from the phase 1 and phase 2, stratum 1 study which included participants with clinically significant PN-related morbidity. Participants received continuous selumetinib dosing (1 cycle = 28 days). Safety and efficacy data through February 27, 2021 are included. PN response assessed by volumetric magnetic resonance imaging analysis: Confirmed partial response (cPR) ≥20% decrease from baseline on 2 consecutive evaluations. Phase 2 participants completed patient-reported outcome measures assessing tumor pain intensity (Numeric Rating Scale-11) and interference of pain in daily life (pain interference index). RESULTS For the 74 children (median age 10.3 years; range 3-18.5) enrolled, overall cPR rate was 70% (52/74); median duration of treatment was 57.5 cycles (range 1-100). Responses were generally sustained with 59% (44) lasting ≥ 12 cycles. Tumor pain intensity (n = 19, P = .015) and pain interference (n = 18, P = .0059) showed durable improvement from baseline to 48 cycles. No new safety signals were identified; however, some developed known selumetinib-related adverse events (AEs) for the first time after several years of treatment. CONCLUSIONS With up to 5 years of additional selumetinib treatment, most children with NF1-related PN had durable tumor shrinkage and sustained improvement in pain beyond that previously reported at 1 year. No new safety signals were identified; however, ongoing monitoring for known selumetinib-related AEs is needed while treatment continues.
Collapse
Affiliation(s)
- Andrea M Gross
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Eva Dombi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Pamela L Wolters
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Andrea Baldwin
- Leidos, Clinical Research Directorate (CRD), Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Anne Dufek
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kailey Herrera
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Staci Martin
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Joanne Derdak
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kara S Heisey
- Leidos, Clinical Research Directorate (CRD), Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Patricia M Whitcomb
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - David J Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Michael J Fisher
- Children’s Hospital of Philadelphia, Section of Neuro-Oncology, Philadelphia, Pennsylvania, USA
| | - AeRang Kim
- Children’s National Hospital, Center for Cancer and Blood Disorders, Washington, District of Columbia, USA
| | - Miriam Bornhorst
- Children’s National Hospital, Center for Cancer and Blood Disorders, Washington, District of Columbia, USA
| | - Brian D Weiss
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jaishri O Blakeley
- Johns Hopkins University, Division of Neurology, Baltimore, Maryland, USA
| | - Malcolm A Smith
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Brigitte C Widemann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Mezi S, Botticelli A, Scagnoli S, Pomati G, Fiscon G, De Galitiis F, Di Pietro FR, Verkhovskaia S, Amirhassankhani S, Pisegna S, Gentile G, Simmaco M, Gohlke B, Preissner R, Marchetti P. The Impact of Drug-Drug Interactions on the Toxicity Profile of Combined Treatment with BRAF and MEK Inhibitors in Patients with BRAF-Mutated Metastatic Melanoma. Cancers (Basel) 2023; 15:4587. [PMID: 37760556 PMCID: PMC10526382 DOI: 10.3390/cancers15184587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND BRAF and MEK inhibition is a successful strategy in managing BRAF-mutant melanoma, even if the treatment-related toxicity is substantial. We analyzed the role of drug-drug interactions (DDI) on the toxicity profile of anti-BRAF/anti-MEK therapy. METHODS In this multicenter, observational, and retrospective study, DDIs were assessed using Drug-PIN software (V 2/23). The association between the Drug-PIN continuous score or the Drug-PIN traffic light and the occurrence of treatment-related toxicities and oncological outcomes was evaluated. RESULTS In total, 177 patients with advanced BRAF-mutated melanoma undergoing BRAF/MEK targeted therapy were included. All grade toxicity was registered in 79% of patients. Cardiovascular toxicities occurred in 31 patients (17.5%). Further, 94 (55.9%) patients had comorbidities requiring specific pharmacological treatments. The median Drug-PIN score significantly increased when the target combination was added to the patient's home therapy (p-value < 0.0001). Cardiovascular toxicity was significantly associated with the Drug-PIN score (p-value = 0.048). The Drug-PIN traffic light (p = 0.00821) and the Drug-PIN score (p = 0.0291) were seen to be significant predictors of cardiotoxicity. Patients with low-grade vs. high-grade interactions showed a better prognosis regarding overall survival (OS) (p = 0.0045) and progression-free survival (PFS) (p = 0.012). The survival analysis of the subgroup of patients with cardiological toxicity demonstrated that patients with low-grade vs. high-grade DDIs had better outcomes in terms of OS (p = 0.0012) and a trend toward significance in PFS (p = 0.068). CONCLUSIONS DDIs emerged as a critical issue for the risk of treatment-related cardiovascular toxicity. Our findings support the utility of DDI assessment in melanoma patients treated with BRAF/MEK inhibitors.
Collapse
Affiliation(s)
- Silvia Mezi
- Department of Radiological, Oncological, and Anatomopathological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (A.B.)
| | - Andrea Botticelli
- Department of Radiological, Oncological, and Anatomopathological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (A.B.)
| | - Simone Scagnoli
- Department of Radiological, Oncological, and Anatomopathological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (A.B.)
| | - Giulia Pomati
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.P.); (S.P.)
| | - Giulia Fiscon
- Department of Computer, Control, and Management Engineering “Antonio Ruberti”, Sapienza University of Rome, 00161 Rome, Italy;
| | - Federica De Galitiis
- Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00144 Rome, Italy; (F.D.G.); (F.R.D.P.); (S.V.); (P.M.)
| | - Francesca Romana Di Pietro
- Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00144 Rome, Italy; (F.D.G.); (F.R.D.P.); (S.V.); (P.M.)
| | - Sofia Verkhovskaia
- Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00144 Rome, Italy; (F.D.G.); (F.R.D.P.); (S.V.); (P.M.)
| | - Sasan Amirhassankhani
- Department of Urology, S. Orsola-Malpighi Hospital, University of Bologna, Via Palagi, 40126 Bologna, Italy;
| | - Simona Pisegna
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.P.); (S.P.)
| | - Giovanna Gentile
- Department of Neuroscience, Mental Health, and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00185 Rome, Italy; (G.G.); (M.S.)
- Unit of Laboratory and Advanced Molecular Diagnostics, ‘Sant’Andrea’ University Hospital, 00189 Rome, Italy
| | - Maurizio Simmaco
- Department of Neuroscience, Mental Health, and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00185 Rome, Italy; (G.G.); (M.S.)
- Unit of Laboratory and Advanced Molecular Diagnostics, ‘Sant’Andrea’ University Hospital, 00189 Rome, Italy
| | - Bjoern Gohlke
- Structural Bioinformatics Group, Institute for Physiology, Charité-University Medicine Berlin, 10117 Berlin, Germany; (B.G.); (R.P.)
| | - Robert Preissner
- Structural Bioinformatics Group, Institute for Physiology, Charité-University Medicine Berlin, 10117 Berlin, Germany; (B.G.); (R.P.)
| | - Paolo Marchetti
- Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00144 Rome, Italy; (F.D.G.); (F.R.D.P.); (S.V.); (P.M.)
| |
Collapse
|
11
|
Marani A, Gioacchini H, Paolinelli M, Offidani A, Campanati A. Potential drug-drug interactions with mitogen-activated protein kinase (MEK) inhibitors used to treat melanoma. Expert Opin Drug Metab Toxicol 2023; 19:555-567. [PMID: 37659065 DOI: 10.1080/17425255.2023.2255519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/08/2023] [Accepted: 09/01/2023] [Indexed: 09/05/2023]
Abstract
INTRODUCTION The management of patients with BRAF-mutated advanced melanoma who are undergoing targeted therapy with MEK inhibitors can be complicated by the co-administration of multiple medications, which can give rise to drug-drug interactions of clinical significance. COVERED AREAS Our review presents a comprehensive analysis of the pharmacokinetic and pharmacodynamic interactions of the three approved for advanced melanoma MEK inhibitor drugs - binimetinib, cobimetinib, and trametinib. MEDLINE (PubMed) was utilized for the literature search, comprising clinical studies, observational studies, and preclinical research. The review discusses the impact of these interactions on efficacy and safety of the treatments and differentiates between interactions supported by pharmacokinetic or pharmacodynamic mechanisms, those encountered in clinical practice, and those observed in preclinical studies. EXPERT OPINION Physicians should be aware about potential benefits, but also increased toxicity caused by drug interactions between MEK inhibitors and other drugs in the management of patients with metastatic melanoma.
Collapse
Affiliation(s)
- A Marani
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - H Gioacchini
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - M Paolinelli
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - A Offidani
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - A Campanati
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| |
Collapse
|
12
|
Butel-Simoes LE, Haw TJ, Williams T, Sritharan S, Gadre P, Herrmann SM, Herrmann J, Ngo DTM, Sverdlov AL. Established and Emerging Cancer Therapies and Cardiovascular System: Focus on Hypertension-Mechanisms and Mitigation. Hypertension 2023; 80:685-710. [PMID: 36756872 PMCID: PMC10023512 DOI: 10.1161/hypertensionaha.122.17947] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Cardiovascular disease and cancer are 2 of the leading causes of death worldwide. Although improvements in outcomes have been noted for both disease entities, the success of cancer therapies has come at the cost of at times very impactful adverse events such as cardiovascular events. Hypertension has been noted as both, a side effect as well as a risk factor for the cardiotoxicity of cancer therapies. Some of these dynamics are in keeping with the role of hypertension as a cardiovascular risk factor not only for heart failure, but also for the development of coronary and cerebrovascular disease, and kidney disease and its association with a higher morbidity and mortality overall. Other aspects such as the molecular mechanisms underlying the amplification of acute and long-term cardiotoxicity risk of anthracyclines and increase in blood pressure with various cancer therapeutics remain to be elucidated. In this review, we cover the latest clinical data regarding the risk of hypertension across a spectrum of novel anticancer therapies as well as the underlying known or postulated pathophysiological mechanisms. Furthermore, we review the acute and long-term implications for the amplification of the development of cardiotoxicity with drugs not commonly associated with hypertension such as anthracyclines. An outline of management strategies, including pharmacological and lifestyle interventions as well as models of care aimed to facilitate early detection and more timely management of hypertension in patients with cancer and survivors concludes this review, which overall aims to improve both cardiovascular and cancer-specific outcomes.
Collapse
Affiliation(s)
- Lloyd E Butel-Simoes
- Cardiovascular Department, John Hunter Hospital, Newcastle, NSW, Australia
- College of Health and Medicine, University of Newcastle, NSW Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW Australia
| | - Tatt Jhong Haw
- College of Health and Medicine, University of Newcastle, NSW Australia
- Newcastle Centre of Excellence in Cardio-Oncology, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW Australia
| | - Trent Williams
- College of Health and Medicine, University of Newcastle, NSW Australia
- Newcastle Centre of Excellence in Cardio-Oncology, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW Australia
| | - Shanathan Sritharan
- Department of Medicine, Hunter New England Local Health District, NSW, Australia
| | - Payal Gadre
- Department of Medicine, Hunter New England Local Health District, NSW, Australia
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55902, USA
| | - Doan TM Ngo
- College of Health and Medicine, University of Newcastle, NSW Australia
- Newcastle Centre of Excellence in Cardio-Oncology, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW Australia
| | - Aaron L Sverdlov
- Cardiovascular Department, John Hunter Hospital, Newcastle, NSW, Australia
- College of Health and Medicine, University of Newcastle, NSW Australia
- Newcastle Centre of Excellence in Cardio-Oncology, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW Australia
| |
Collapse
|
13
|
Wang C, Wang H, Zheng C, Li B, Liu Z, Zhang L, Yuan L, Xu P. Discovery of Coumarin-Based MEK1/2 PROTAC Effective in Human Cancer Cells. ACS Med Chem Lett 2023; 14:92-102. [PMID: 36655129 PMCID: PMC9841598 DOI: 10.1021/acsmedchemlett.2c00446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
The RAF/MEK/ERK pathway is a crucial signal path which is closely associated with the proliferation, differentiation, and apoptosis of tumors. MEK1/2 is a key kinase target in the pathway, with ERK1/2 acting as the main substrate of it. Despite the rapid development of MEK1/2 inhibitors, acquired resistance still happens and remains a significant problem. Most of the inhibitors possess a similar diarylamine scaffold. Here we designed and synthesized a series of MEK1/2 degraders based on a coumarin derivative which was a potent non-diarylamine allosteric MEK1/2 inhibitor. P6b among them showed the most potent degradation effect, with DC50 values of 0.3 μM and 0.2 μM in MEK1 and MEK2 degradation, respectively. An antiproliferation assay showed that it more significantly inhibits the growth of A375 cells (IC50= 2.8 μM) compared to A549 cells (IC50 = 27.3 μM). To sum up, we discovered P6b with a non-diarylamine scaffold for the first time as a potent MEK PROTAC effective in human cancer cells.
Collapse
Affiliation(s)
- Chao Wang
- National
Pharmaceutical Teaching Laboratory Center, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
| | - Han Wang
- Department
of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Cangxin Zheng
- Department
of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bingru Li
- State
Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Zhenming Liu
- State
Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Liangren Zhang
- State
Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Lan Yuan
- Peking
University Medical and Health Analysis Center, Peking University Health Science Center, Beijing 100191, China
| | - Ping Xu
- Department
of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
14
|
Di Nunno V, Gatto L, Tosoni A, Bartolini S, Franceschi E. Implications of BRAF V600E mutation in gliomas: Molecular considerations, prognostic value and treatment evolution. Front Oncol 2023; 12:1067252. [PMID: 36686797 PMCID: PMC9846085 DOI: 10.3389/fonc.2022.1067252] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
Gliomas are molecularly heterogeneous brain tumors responsible for the most years of life lost by any cancer. High-grade gliomas have a poor prognosis and despite multimodal treatment including surgery, radiotherapy, and chemotherapy, exhibit a high recurrence rate. There is a need for new therapeutic approaches based on precision medicine informed by biomarker assessment and BRAF, a key regulator of MAPK signaling pathway, influencing cell differentiation, proliferation, migration and pro-tumorigenic activity, is emerging as a promising molecular target. V600E, is the most frequent BRAF alteration in gliomas, especially in pediatric low-grade astrocytomas, pleomorphic xanthoastrocytoma, papillary craniopharyngioma, epithelioid glioblastoma and ganglioglioma. The possible application of BRAF-targeted therapy in gliomas is continuously growing and there is preliminary evidence of prolonged disease control obtained by BRAF inhibitors in tumors harboring BRAF V600E mutation. The possibility of introducing targeted therapies into the treatment algorithm represents a paradigm shift for patients with BRAF V600E mutant recurrent high-grade and low-grade glioma and BRAF routine testing should be considered in clinical practice. The focus of this review is to summarize the molecular landscape of BRAF across glioma subtypes and the novel therapeutic strategies for BRAF V600E mutated tumors.
Collapse
Affiliation(s)
| | - Lidia Gatto
- Department of Oncology, AUSL Bologna, Bologna, Italy,*Correspondence: Lidia Gatto,
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
15
|
Wu X, Shen F, Jiang G, Xue G, Philips S, Gardner L, Cunningham G, Bales C, Cantor E, Schneider BP. A non-coding GWAS variant impacts anthracycline-induced cardiotoxic phenotypes in human iPSC-derived cardiomyocytes. Nat Commun 2022; 13:7171. [PMID: 36418322 PMCID: PMC9684507 DOI: 10.1038/s41467-022-34917-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/11/2022] [Indexed: 11/24/2022] Open
Abstract
Anthracyclines, widely used to treat breast cancer, have the potential for cardiotoxicity. We have previously identified and validated a germline single nucleotide polymorphism, rs28714259, associated with an increased risk of anthracycline-induced heart failure. We now provide insights into the mechanism by which rs28714259 might confer increased risk of cardiac damage. Using hiPSC-derived cardiomyocyte cell lines with either intrinsic polymorphism or CRISPR-Cas9-mediated deletion of rs28714259 locus, we demonstrate that glucocorticoid receptor signaling activated by dexamethasone pretreatment prior to doxorubicin exposure preserves cardiomyocyte viability and contractility in cardiomyocytes containing the major allele. Homozygous loss of the rs28714259 major allele diminishes dexamethasone's protective effect. We further demonstrate that the risk allele of rs28714259 disrupts glucocorticoid receptor and rs28714259 binding affinity. Finally, we highlight the activation of genes and pathways involved in cardiac hypertrophy signaling that are blocked by the risk allele, suggesting a decreased adaptive survival response to doxorubicin-related stress.
Collapse
Affiliation(s)
- Xi Wu
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fei Shen
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Guanglong Jiang
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Gloria Xue
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Santosh Philips
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Laura Gardner
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Geneva Cunningham
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Casey Bales
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Erica Cantor
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bryan Paul Schneider
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
16
|
Lyon AR, López-Fernández T, Couch LS, Asteggiano R, Aznar MC, Bergler-Klein J, Boriani G, Cardinale D, Cordoba R, Cosyns B, Cutter DJ, de Azambuja E, de Boer RA, Dent SF, Farmakis D, Gevaert SA, Gorog DA, Herrmann J, Lenihan D, Moslehi J, Moura B, Salinger SS, Stephens R, Suter TM, Szmit S, Tamargo J, Thavendiranathan P, Tocchetti CG, van der Meer P, van der Pal HJH. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur Heart J 2022; 43:4229-4361. [PMID: 36017568 DOI: 10.1093/eurheartj/ehac244] [Citation(s) in RCA: 886] [Impact Index Per Article: 443.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
17
|
Beck TC, Arhontoulis DC, Morningstar JE, Hyams N, Stoddard A, Springs K, Mukherjee R, Helke K, Guo L, Moore K, Gensemer C, Biggs R, Petrucci T, Kwon J, Stayer K, Koren N, Harvey A, Holman H, Dunne J, Fulmer D, Vohra A, Mai L, Dooley S, Weninger J, Vaena S, Romeo M, Muise-Helmericks RC, Mei Y, Norris RA. Cellular and Molecular Mechanisms of MEK1 Inhibitor-Induced Cardiotoxicity. JACC CardioOncol 2022; 4:535-548. [PMID: 36444237 PMCID: PMC9700254 DOI: 10.1016/j.jaccao.2022.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Trametinib is a MEK1 (mitogen-activated extracellular signal-related kinase kinase 1) inhibitor used in the treatment of BRAF (rapid accelerated fibrosarcoma B-type)-mutated metastatic melanoma. Roughly 11% of patients develop cardiomyopathy following long-term trametinib exposure. Although described clinically, the molecular landscape of trametinib cardiotoxicity has not been characterized. Objectives The aim of this study was to test the hypothesis that trametinib promotes widespread transcriptomic and cellular changes consistent with oxidative stress and impairs cardiac function. Methods Mice were treated with trametinib (1 mg/kg/d). Echocardiography was performed pre- and post-treatment. Gross, histopathologic, and biochemical assessments were performed to probe for molecular and cellular changes. Human cardiac organoids were used as an in vitro measurement of cardiotoxicity and recovery. Results Long-term administration of trametinib was associated with significant reductions in survival and left ventricular ejection fraction. Histologic analyses of the heart revealed myocardial vacuolization and calcification in 28% of animals. Bulk RNA sequencing identified 435 differentially expressed genes and 116 differential signaling pathways following trametinib treatment. Upstream gene analysis predicted interleukin-6 as a regulator of 17 relevant differentially expressed genes, suggestive of PI3K/AKT and JAK/STAT activation, which was subsequently validated. Trametinib hearts displayed elevated markers of oxidative stress, myofibrillar degeneration, an 11-fold down-regulation of the apelin receptor, and connexin-43 mislocalization. To confirm the direct cardiotoxic effects of trametinib, human cardiac organoids were treated for 6 days, followed by a 6-day media-only recovery. Trametinib-treated organoids exhibited reductions in diameter and contractility, followed by partial recovery with removal of treatment. Conclusions These data describe pathologic changes observed in trametinib cardiotoxicity, supporting the exploration of drug holidays and alternative pharmacologic strategies for disease prevention.
Collapse
Affiliation(s)
- Tyler C. Beck
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Dimitrios C. Arhontoulis
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Jordan E. Morningstar
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Nathaniel Hyams
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Andrew Stoddard
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kendra Springs
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Rupak Mukherjee
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kris Helke
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lilong Guo
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kelsey Moore
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Cortney Gensemer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Rachel Biggs
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Taylor Petrucci
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jennie Kwon
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kristina Stayer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Natalie Koren
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Andrew Harvey
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Heather Holman
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jaclyn Dunne
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Diana Fulmer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ayesha Vohra
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Le Mai
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sarah Dooley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Julianna Weninger
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Silvia Vaena
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Martin Romeo
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Robin C. Muise-Helmericks
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ying Mei
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Russell A. Norris
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
18
|
Lyon AR, López-Fernández T, Couch LS, Asteggiano R, Aznar MC, Bergler-Klein J, Boriani G, Cardinale D, Cordoba R, Cosyns B, Cutter DJ, de Azambuja E, de Boer RA, Dent SF, Farmakis D, Gevaert SA, Gorog DA, Herrmann J, Lenihan D, Moslehi J, Moura B, Salinger SS, Stephens R, Suter TM, Szmit S, Tamargo J, Thavendiranathan P, Tocchetti CG, van der Meer P, van der Pal HJH. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur Heart J Cardiovasc Imaging 2022; 23:e333-e465. [PMID: 36017575 DOI: 10.1093/ehjci/jeac106] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
19
|
de Wit S, Glen C, de Boer RA, Lang NN. Mechanisms shared between cancer, heart failure, and targeted anti-cancer therapies. Cardiovasc Res 2022; 118:3451-3466. [PMID: 36004495 PMCID: PMC9897696 DOI: 10.1093/cvr/cvac132] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/12/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023] Open
Abstract
Heart failure (HF) and cancer are the leading causes of death worldwide and accumulating evidence demonstrates that HF and cancer affect one another in a bidirectional way. Patients with HF are at increased risk for developing cancer, and HF is associated with accelerated tumour growth. The presence of malignancy may induce systemic metabolic, inflammatory, and microbial alterations resulting in impaired cardiac function. In addition to pathophysiologic mechanisms that are shared between cancer and HF, overlaps also exist between pathways required for normal cardiac physiology and for tumour growth. Therefore, these overlaps may also explain the increased risk for cardiotoxicity and HF as a result of targeted anti-cancer therapies. This review provides an overview of mechanisms involved in the bidirectional connection between HF and cancer, specifically focusing upon current 'hot-topics' in these shared mechanisms. It subsequently describes targeted anti-cancer therapies with cardiotoxic potential as a result of overlap between their anti-cancer targets and pathways required for normal cardiac function.
Collapse
Affiliation(s)
- Sanne de Wit
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, PO Box 30.001, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Claire Glen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, PO Box 30.001, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | | |
Collapse
|
20
|
Elnaggar M, Agte S, Restrepo P, Ram M, Melnekoff D, Adamopoulos C, Stevens MM, Kappes K, Leshchenko V, Verina D, Jagannath S, Poulikakos PI, Parekh S, Laganà A. Triple MAPK inhibition salvaged a relapsed post-BCMA CAR-T cell therapy multiple myeloma patient with a BRAF V600E subclonal mutation. J Hematol Oncol 2022; 15:109. [PMID: 35978321 PMCID: PMC9382834 DOI: 10.1186/s13045-022-01330-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/05/2022] [Indexed: 11/18/2022] Open
Abstract
Background Multiple Myeloma (MM) is a progressive plasma cell neoplasm characterized by heterogeneous clonal expansion. Despite promising response rates achieved with anti-BCMA CAR-T cell therapy, patients may still relapse and there are currently no clear therapeutic options in post-CAR-T settings. In this report, we present a case of a post-BCMA CAR-T relapsed/refractory (RR) MM patient with skin extramedullary disease (EMD) in which a novel MAPK inhibition combinatorial strategy was implemented based on next-generation sequencing and in vitro experiments. Case presentation A 61-year-old male with penta-refractory MM penta- (IgA lambda), ISS stage 3 with hyperdiploidy, gain of 1q21 and del13 was treated with anti-BCMA CAR-T cell therapy, achieving a best response of VGPR. He progressed after 6 months and was salvaged for a short period with autologous stem cell transplantation. Eventually, he progressed with extramedullary disease manifested as subcutaneous nodules. Based on whole-exome sequencing, we identified a BRAF (V600E) dominant subclone in both bone marrow and cutaneous plasmacytoma. Following in vitro experiments, and according to our previous studies, we implemented a triple MAPK inhibition strategy under which the patient achieved a very good partial response for 110 days, which allowed to bridge him to subsequent clinical trials and eventually achieve a stringent complete response (sCR). Conclusion Here, we show the applicability, effectiveness, and tolerability the triple MAPK inhibition strategy in the context of post-BCMA CAR-T failure in specific subset of patients. The triple therapy could bridge our hospice bound RRMM patient with BRAF (V600E) to further therapeutic options where sCR was achieved. We will further evaluate triple MAPK inhibition in patients with BRAF V600E in a precision medicine clinical trial launching soon. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01330-3.
Collapse
Affiliation(s)
- Muhammad Elnaggar
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarita Agte
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paula Restrepo
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meghana Ram
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Melnekoff
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christos Adamopoulos
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Katerina Kappes
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Violetta Leshchenko
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Verina
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sundar Jagannath
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Poulikos I Poulikakos
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samir Parekh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alessandro Laganà
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
21
|
Robison NJ, Su JA, Fang MJ, Malvar J, Menteer J. Cardiac Function in Children and Young Adults Treated with MEK Inhibitors: A Retrospective Cohort Study. Pediatr Cardiol 2022; 43:1223-1228. [PMID: 35233653 PMCID: PMC10284303 DOI: 10.1007/s00246-022-02842-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/03/2022] [Indexed: 11/26/2022]
Abstract
MEK inhibitors (MEKi) have shown efficacy in pediatric low-grade glioma as well as plexiform neurofibroma. MEKi have been associated with acute cardiac dysfunction in adults. Cardiac consequences in children are unknown. We performed a single center retrospective cohort study evaluating cardiac function by echocardiography (echo) in children and young adults < 21 years receiving MEKi between October 2013 and May 2018. Blinded assessment of left ventricular function by fractional shortening (FS) and ejection fraction (EF) was performed on all available echocardiograms performed before, during, and following therapy, as well as after re-initiation of therapy. Twenty-six patients underwent MEKi therapy with echo follow-up during the study period. Twenty-four of these had complete echo data. Median follow-up was 12 months. Borderline EF (EF 53-57.9%) occurred in 12 (50%) patients; and 3 (12.5%) progressed to abnormal EF (EF < 53%). Cardiac dysfunction, when it occurred, was mild (lowest documented EF was 45%, and lowest FS was 24.4%). EF abnormalities typically fluctuated during therapy, resolved off therapy, and recurred with MEKi re-initiation. No clinical or demographic differences were detected between those who maintained normal cardiac function and those who developed borderline or overt cardiac dysfunction. Symptomatic heart failure did not occur. In this cohort of children and young adults, MEKi use was associated with a high (50%) incidence of borderline or mildly decreased left ventricular function. There was no evidence of permanent cardiac dysfunction. Further evaluation in larger prospective trials is needed.
Collapse
Affiliation(s)
- Nathan J Robison
- Division of Hematology and Oncology, Children's Hospital Los Angeles, 4650 W. Sunset Blvd, MS#54, Los Angeles, CA, 90027, USA.
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Jennifer A Su
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Cardiology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Melody J Fang
- Chicago Medical School at Rosalind Franklin University, North Chicago, IL, USA
| | - Jemily Malvar
- Division of Hematology and Oncology, Children's Hospital Los Angeles, 4650 W. Sunset Blvd, MS#54, Los Angeles, CA, 90027, USA
| | - Jondavid Menteer
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Cardiology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
22
|
Wang CY, Zoungas S, Voskoboynik M, Mar V. Cardiovascular disease and malignant melanoma. Melanoma Res 2022; 32:135-141. [PMID: 35377865 DOI: 10.1097/cmr.0000000000000817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In the last decade, systemic therapies such as immune checkpoint inhibitors and BRAF-MEK inhibitors have improved the prognosis of high-risk and advanced melanoma. With improved survival, melanoma survivorship is increasingly important, particularly in patients who have a good prognosis or are diagnosed at a younger age. It is increasingly recognized that cancer and its treatment is associated with increased cardiovascular morbidity and mortality. Indeed, data from observational studies and meta-analyses of randomized controls trials in melanoma show that systemic therapies may be associated with cardiac toxicities, such as myocardial infarction, heart failure, myocarditis and stroke. Our review will discuss cardiovascular disease and risk factors in the context of melanoma and outline the importance of cardiovascular risk modification in this population.
Collapse
Affiliation(s)
- Charlie Yue Wang
- Victorian Melanoma Service, The Alfred Hospital
- School of Public Health and Preventative Medicine, Monash University
| | - Sophia Zoungas
- School of Public Health and Preventative Medicine, Monash University
| | - Mark Voskoboynik
- Department of Oncology, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Victoria Mar
- Victorian Melanoma Service, The Alfred Hospital
- School of Public Health and Preventative Medicine, Monash University
| |
Collapse
|
23
|
Meilhac A, Cautela J, Thuny F. Cancer Therapies and Vascular Toxicities. Curr Treat Options Oncol 2022; 23:333-347. [PMID: 35244888 DOI: 10.1007/s11864-022-00964-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 12/24/2022]
Abstract
OPINION STATEMENT Vascular events have become an important issue in the overall management of cancer patients. They usually result from a combination of (i) direct or indirect toxicity of anticancer treatments, (ii) a higher prevalence of cardiovascular risk factors in cancer patients, and (iii) prolonged exposure to treatments due to an increasing patient survival rate. In addition to conventional chemotherapies and radiotherapy, targeted therapies and immunotherapies have been developed which improve the prognosis of cancer patients but sometimes at the cost of vascular toxicity, which can lead to systemic or pulmonary hypertension and arterial/venous thromboembolic events. Endothelial dysfunction, a procoagulant state and metabolic disorders are the three main pathophysiological patterns leading to cancer treatment-related vascular toxicity. This issue is challenging because serious vascular adverse events can necessitate cancer treatment being put on hold or stopped, which could compromise patient survival. In addition to increasing the risk of thrombotic adverse events, cancer therapies may lead to an increased risk of bleeding, especially in treatments with vascular endothelial growth factor inhibitors. Therefore, we can define vasculo-oncology as a part of the cardio-oncology specialty; its aims are to predict, prevent, screen, and treat vascular toxicity related to cancer treatments. While the level of evidence is low regarding the management of vascular toxicity during cancer therapy, cardiologists and specialists in vascular diseases should closely collaborate with oncologists and hematologists to determine the optimal strategy for each patient.
Collapse
Affiliation(s)
- Alexandra Meilhac
- Department of Cardiology, North Hospital, Assistance Publique - Hôpitaux de Marseille, Centre for CardioVascular and Nutrition Research (C2VN), University Mediterranean Center of Cardio-Oncology, Unit of Heart Failure and Valvular Heart Diseases, Inserm 1263, Inrae 1260, Aix-Marseille University, Chemin des Bourrely, 13015, Marseille, France
| | - Jennifer Cautela
- Department of Cardiology, North Hospital, Assistance Publique - Hôpitaux de Marseille, Centre for CardioVascular and Nutrition Research (C2VN), University Mediterranean Center of Cardio-Oncology, Unit of Heart Failure and Valvular Heart Diseases, Inserm 1263, Inrae 1260, Aix-Marseille University, Chemin des Bourrely, 13015, Marseille, France
| | - Franck Thuny
- Department of Cardiology, North Hospital, Assistance Publique - Hôpitaux de Marseille, Centre for CardioVascular and Nutrition Research (C2VN), University Mediterranean Center of Cardio-Oncology, Unit of Heart Failure and Valvular Heart Diseases, Inserm 1263, Inrae 1260, Aix-Marseille University, Chemin des Bourrely, 13015, Marseille, France.
| |
Collapse
|
24
|
Grela-Wojewoda A, Pacholczak-Madej R, Adamczyk A, Korman M, Püsküllüoğlu M. Cardiotoxicity Induced by Protein Kinase Inhibitors in Patients with Cancer. Int J Mol Sci 2022; 23:ijms23052815. [PMID: 35269958 PMCID: PMC8910876 DOI: 10.3390/ijms23052815] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 12/24/2022] Open
Abstract
Kinase inhibitors (KIs) represent a growing class of drugs directed at various protein kinases and used in the treatment of both solid tumors and hematologic malignancies. It is a heterogeneous group of compounds that are widely applied not only in different types of tumors but also in tumors that are positive for a specific predictive factor. This review summarizes common cardiotoxic effects of KIs, including hypertension, arrhythmias with bradycardia and QTc prolongation, and cardiomyopathy that can lead to heart failure, as well as less common effects such as fluid retention, ischemic heart disease, and elevated risk of thromboembolic events. The guidelines for cardiac monitoring and management of the most common cardiotoxic effects of protein KIs are discussed. Potential signaling pathways affected by KIs and likely contributing to cardiac damage are also described. Finally, the need for further research into the molecular mechanisms underlying the cardiovascular toxicity of these drugs is indicated.
Collapse
Affiliation(s)
- Aleksandra Grela-Wojewoda
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
- Correspondence: ; Tel.: +48-1263-48350
| | - Renata Pacholczak-Madej
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
- Department of Anatomy, Jagiellonian University Medical College, 31-008 Kraków, Poland
| | - Agnieszka Adamczyk
- Department of Tumour Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland;
| | - Michał Korman
- Faculty of Medicine, Jagiellonian University Medical College, 31-008 Kraków, Poland;
| | - Mirosława Püsküllüoğlu
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
| |
Collapse
|
25
|
Glen C, Tan YY, Waterston A, Evans TRJ, Jones RJ, Petrie MC, Lang NN. Mechanistic and Clinical Overview Cardiovascular Toxicity of BRAF and MEK Inhibitors: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2022; 4:1-18. [PMID: 35492830 PMCID: PMC9040125 DOI: 10.1016/j.jaccao.2022.01.096] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/07/2022] [Indexed: 02/07/2023] Open
Abstract
Rapidly accelerated fibrosarcoma B-type (BRAF) and mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors have revolutionized melanoma treatment. Approximately half of patients with melanoma harbor a BRAF gene mutation with subsequent dysregulation of the RAF-MEK-ERK signaling pathway. Targeting this pathway with BRAF and MEK blockade results in control of cell proliferation and, in most cases, disease control. These pathways also have cardioprotective effects and are necessary for normal vascular and cardiac physiology. BRAF and MEK inhibitors are associated with adverse cardiovascular effects including hypertension, left ventricular dysfunction, venous thromboembolism, atrial arrhythmia, and electrocardiographic QT interval prolongation. These effects may be underestimated in clinical trials. Baseline cardiovascular assessment and follow-up, including serial imaging and blood pressure assessment, are essential to balance optimal anti-cancer therapy while minimizing cardiovascular side effects. In this review, an overview of BRAF/MEK inhibitor-induced cardiovascular toxicity, the mechanisms underlying these, and strategies for surveillance, prevention, and treatment of these effects are provided.
Collapse
Key Words
- ACE, angiotensin-converting enzyme
- AF, atrial fibrillation
- BRAF inhibitor
- BRAF, rapidly accelerated fibrosarcoma B-type
- CVAE, cardiovascular adverse event
- EGFR, epidermal growth factor receptor
- ERK, extracellular signal-regulated kinase
- LVSD, left ventricular systolic dysfunction
- MEK inhibitor
- MEK, mitogen-activated extracellular signal-regulated kinase
- RAF, rapidly accelerated fibrosarcoma
- VEGF, vascular endothelial growth factor
- cardio-oncology
- cardiovascular toxicity
- hypertension
- left ventricular systolic dysfunction
- melanoma
Collapse
Affiliation(s)
- Claire Glen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Yun Yi Tan
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Ashita Waterston
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Thomas R. Jeffry Evans
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Robert J. Jones
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mark C. Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ninian N. Lang
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| |
Collapse
|
26
|
Clerk A, Meijles DN, Hardyman MA, Fuller SJ, Chothani SP, Cull JJ, Cooper ST, Alharbi HO, Vanezis K, Felkin LE, Markou T, Leonard SJ, Shaw SW, Rackham OJ, Cook SA, Glennon PE, Sheppard MN, Sembrat JC, Rojas M, McTiernan CF, Barton PJ, Sugden PH. Cardiomyocyte BRAF and type 1 RAF inhibitors promote cardiomyocyte and cardiac hypertrophy in mice in vivo. Biochem J 2022; 479:401-424. [PMID: 35147166 PMCID: PMC8883496 DOI: 10.1042/bcj20210615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/12/2022] [Accepted: 01/27/2022] [Indexed: 12/13/2022]
Abstract
The extracellular signal-regulated kinase 1/2 (ERK1/2) cascade promotes cardiomyocyte hypertrophy and is cardioprotective, with the three RAF kinases forming a node for signal integration. Our aims were to determine if BRAF is relevant for human heart failure, whether BRAF promotes cardiomyocyte hypertrophy, and if Type 1 RAF inhibitors developed for cancer (that paradoxically activate ERK1/2 at low concentrations: the 'RAF paradox') may have the same effect. BRAF was up-regulated in heart samples from patients with heart failure compared with normal controls. We assessed the effects of activated BRAF in the heart using mice with tamoxifen-activated Cre for cardiomyocyte-specific knock-in of the activating V600E mutation into the endogenous gene. We used echocardiography to measure cardiac dimensions/function. Cardiomyocyte BRAFV600E induced cardiac hypertrophy within 10 d, resulting in increased ejection fraction and fractional shortening over 6 weeks. This was associated with increased cardiomyocyte size without significant fibrosis, consistent with compensated hypertrophy. The experimental Type 1 RAF inhibitor, SB590885, and/or encorafenib (a RAF inhibitor used clinically) increased ERK1/2 phosphorylation in cardiomyocytes, and promoted hypertrophy, consistent with a 'RAF paradox' effect. Both promoted cardiac hypertrophy in mouse hearts in vivo, with increased cardiomyocyte size and no overt fibrosis. In conclusion, BRAF potentially plays an important role in human failing hearts, activation of BRAF is sufficient to induce hypertrophy, and Type 1 RAF inhibitors promote hypertrophy via the 'RAF paradox'. Cardiac hypertrophy resulting from these interventions was not associated with pathological features, suggesting that Type 1 RAF inhibitors may be useful to boost cardiomyocyte function.
Collapse
Affiliation(s)
- Angela Clerk
- School of Biological Sciences, University of Reading, Reading, U.K
| | - Daniel N. Meijles
- Molecular and Clinical Sciences Institute, St. George's University of London, London, U.K
| | | | | | - Sonia P. Chothani
- Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore City, Singapore
| | - Joshua J. Cull
- School of Biological Sciences, University of Reading, Reading, U.K
| | - Susanna T.E. Cooper
- Molecular and Clinical Sciences Institute, St. George's University of London, London, U.K
| | - Hajed O. Alharbi
- School of Biological Sciences, University of Reading, Reading, U.K
| | - Konstantinos Vanezis
- National Heart and Lung Institute, Imperial College London, London, U.K
- MRC London Institute of Medical Sciences, Imperial College London, London, U.K
| | - Leanne E. Felkin
- National Heart and Lung Institute, Imperial College London, London, U.K
- Cardiovascular Research Centre, Royal Brompton and Harefield Hospitals, London, U.K
| | - Thomais Markou
- School of Biological Sciences, University of Reading, Reading, U.K
| | | | - Spencer W. Shaw
- School of Biological Sciences, University of Reading, Reading, U.K
| | - Owen J.L. Rackham
- Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore City, Singapore
| | - Stuart A. Cook
- Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore City, Singapore
- MRC London Institute of Medical Sciences, Imperial College London, London, U.K
- National Heart Centre Singapore, Singapore City, Singapore
| | - Peter E. Glennon
- University Hospitals Coventry and Warwickshire, University Hospital Cardiology Department, Clifford Bridge Road, Coventry, U.K
| | - Mary N. Sheppard
- CRY Cardiovascular Pathology Department, St. George's Healthcare NHS Trust, London, U.K
| | - John C. Sembrat
- Division of Pulmonary, Allergy and Critical Care Medicine, and Dorothy P & Richard P Simmons Center for Interstitial Lung Disease, Department of Medicine, University of Pittsburgh, Pittsburgh, U.S.A
| | - Mauricio Rojas
- Division of Pulmonary, Allergy and Critical Care Medicine, and Dorothy P & Richard P Simmons Center for Interstitial Lung Disease, Department of Medicine, University of Pittsburgh, Pittsburgh, U.S.A
| | - Charles F. McTiernan
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, U.S.A
| | - Paul J. Barton
- National Heart and Lung Institute, Imperial College London, London, U.K
- Cardiovascular Research Centre, Royal Brompton and Harefield Hospitals, London, U.K
| | - Peter H. Sugden
- School of Biological Sciences, University of Reading, Reading, U.K
| |
Collapse
|
27
|
Courand PY, Berger M, Bouali A, Harbaoui B, Lantelme P, Dalle S. Cardiac Effects of BRAF and MEK Inhibitors: Mechanisms and Clinical Management. Curr Oncol Rep 2022; 24:265-271. [PMID: 35102484 DOI: 10.1007/s11912-022-01205-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW The identification of BRAF mutation prompted the development of new class of targeted therapy for treating melanoma: BRAF inhibitors and MEK inhibitors. Cardiovascular events have been reported with these treatments and could counterbalance their long-term maintenance. RECENT FINDINGS LVEF decrease due to BRAF and MEK inhibitors appears fairly common (10%) but usually not severe, without impact on patient outcomes. To date, no treatment options have been tested to prevent or to treat a decrease of LVEF associated with BRAF and MEK inhibitors. QTc prolongation was observed in 3% and arterial hypertension in 20% during treatment but only one-third of cases required a therapeutic change. BRAF and MEK inhibitors have revolutionized the management and the prognosis of melanoma patients. Cardio-oncology units may be useful for a better care of potential cardiac toxicity and particularly to inappropriately avoid discontinuing BRAF and MEK inhibitors.
Collapse
Affiliation(s)
- Pierre-Yves Courand
- Fédération de Cardiologie, Hôpital de La Croix-Rousse Et Hôpital Lyon Sud, Hospices Civils de Lyon, 103 Grande Rue de la Croix-Rousse, 69004, Lyon, France. .,Université de Lyon, CREATIS, CNRS UMR5220, INSERM U1044, INSA-Lyon, Université Claude Bernard Lyon 1, Villeurbanne, France.
| | - Mathilde Berger
- Service de Dermatologie, Hôpital de Lyon Sud, Pierre Bénite, France
| | - Anissa Bouali
- Fédération de Cardiologie, Hôpital de La Croix-Rousse Et Hôpital Lyon Sud, Hospices Civils de Lyon, 103 Grande Rue de la Croix-Rousse, 69004, Lyon, France
| | - Brahim Harbaoui
- Fédération de Cardiologie, Hôpital de La Croix-Rousse Et Hôpital Lyon Sud, Hospices Civils de Lyon, 103 Grande Rue de la Croix-Rousse, 69004, Lyon, France.,Université de Lyon, CREATIS, CNRS UMR5220, INSERM U1044, INSA-Lyon, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Pierre Lantelme
- Fédération de Cardiologie, Hôpital de La Croix-Rousse Et Hôpital Lyon Sud, Hospices Civils de Lyon, 103 Grande Rue de la Croix-Rousse, 69004, Lyon, France.,Université de Lyon, CREATIS, CNRS UMR5220, INSERM U1044, INSA-Lyon, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Stéphane Dalle
- Service de Dermatologie, Hôpital de Lyon Sud, Pierre Bénite, France
| |
Collapse
|
28
|
Vaassen P, Dürr NR, Rosenbaum T. Treatment of Plexiform Neurofibromas with MEK Inhibitors: First Results with a New Therapeutic Option. Neuropediatrics 2022; 53:52-60. [PMID: 34905788 DOI: 10.1055/s-0041-1740549] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Neurofibromatosis type-1 (NF1)-associated plexiform neurofibromas (PN) are peripheral nerve sheath tumors that can significantly affect the quality of life. Until recently, surgery was the only treatment for these tumors. However, in most cases, surgery cannot achieve complete tumor removal and carries a high risk of postoperative deficits. Therefore, the recent approval of the MEK inhibitor selumetinib for the treatment of NF1-associated PN provides a long-awaited novel therapeutic option. Here, we report our experience with MEK inhibitor treatment in 12 pediatric NF1 patients with inoperable symptomatic PN. Eight patients received trametinib (median therapy duration 12.13 months and range 4-29 months), and four patients received selumetinib (median therapy duration 6.25 months and range 4-11 months). Volumetric magnetic resonance imaging (MRI) after 6 months of treatment was available for seven trametinib patients (median tumor volume reduction of 26.5% and range 11.3-55.7%) and two selumetinib patients (21.3% tumor volume reduction in one patient and +3% tumor volume change in the other one). All patients reported clinical benefits such as improved range of motion or reduced disfigurement. Therapy-related adverse events occurred in 58.3% of patients and mainly consisted of skin toxicity, paronychia, and gastrointestinal symptoms. Two patients discontinued trametinib treatment after 14 and 29 months when severe skin toxicity occurred and no further reduction of tumor size was observed. In one patient, discontinuation of therapy resulted in a 27.2% tumor volume increase as demonstrated on volumetric MRI 6 months later. Our data show that MEK inhibition is a novel therapeutic approach for inoperable PN with promising results and a manageable safety profile.
Collapse
Affiliation(s)
- Pia Vaassen
- Department of Pediatrics, Sana Kliniken Duisburg, Duisburg, Germany
| | | | | |
Collapse
|
29
|
Tamargo J, Caballero R, Delpón E. Cancer Chemotherapy-Induced Sinus Bradycardia: A Narrative Review of a Forgotten Adverse Effect of Cardiotoxicity. Drug Saf 2022; 45:101-126. [PMID: 35025085 DOI: 10.1007/s40264-021-01132-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 12/20/2022]
Abstract
Cardiotoxicity is a common adverse effect of anticancer drugs (ACDs), including the so-called targeted drugs, and increases morbidity and mortality in patients with cancer. Attention has focused mainly on ACD-induced heart failure, myocardial ischemia, hypertension, thromboembolism, QT prolongation, and tachyarrhythmias. Yet, although an increasing number of ACDs can produce sinus bradycardia (SB), this proarrhythmic effect remains an underappreciated complication, probably because of its low incidence and severity since most patients are asymptomatic. However, SB merits our interest because its incidence increases with the aging of the population and cancer is an age-related disease and because SB represents a risk factor for QT prolongation. Indeed, several ACDs that produce SB also prolong the QT interval. We reviewed published reports on ACD-induced SB from January 1971 to November 2020 using the PubMed and EMBASE databases. Published reports from clinical trials, case reports, and recent reviews were considered. This review describes the associations between ACDs and SB, their clinical relevance, risk factors, and possible mechanisms of onset and treatment.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain.
| | - Ricardo Caballero
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain
| |
Collapse
|
30
|
Gubbi S, Koch CA, Klubo-Gwiezdzinska J. Peptide Receptor Radionuclide Therapy in Thyroid Cancer. Front Endocrinol (Lausanne) 2022; 13:896287. [PMID: 35712243 PMCID: PMC9197113 DOI: 10.3389/fendo.2022.896287] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/19/2022] [Indexed: 01/03/2023] Open
Abstract
The treatment options that are currently available for management of metastatic, progressive radioactive iodine (RAI)-refractory differentiated thyroid cancers (DTCs), and medullary thyroid cancers (MTCs) are limited. While there are several systemic targeted therapies, such as tyrosine kinase inhibitors, that are being evaluated and implemented in the treatment of these cancers, such therapies are associated with serious, sometimes life-threatening, adverse events. Peptide receptor radionuclide therapy (PRRT) has the potential to be an effective and safe modality for treating patients with somatostatin receptor (SSTR)+ RAI-refractory DTCs and MTCs. MTCs and certain sub-types of RAI-refractory DTCs, such as Hürthle cell cancers which are less responsive to conventional modalities of treatment, have demonstrated a favorable response to treatment with PRRT. While the current literature offers hope for utilization of PRRT in thyroid cancer, several areas of this field remain to be investigated further, especially head-to-head comparisons with other systemic targeted therapies. In this review, we provide a comprehensive outlook on the current translational and clinical data on the use of various PRRTs, including diagnostic utility of somatostatin analogs, theranostic properties of PRRT, and the potential areas for future research.
Collapse
Affiliation(s)
- Sriram Gubbi
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Christian A. Koch
- Department of Medicine, Fox Chase Cancer Center, Philadelphia, PA, United States
- Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Joanna Klubo-Gwiezdzinska
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Joanna Klubo-Gwiezdzinska,
| |
Collapse
|
31
|
Li R, Qi Y, Yuan Q, Xu L, Gao M, Xu Y, Han X, Yin L, Liu C. Protective effects of dioscin against isoproterenol-induced cardiac hypertrophy via adjusting PKCε/ERK-mediated oxidative stress. Eur J Pharmacol 2021; 907:174277. [PMID: 34171391 DOI: 10.1016/j.ejphar.2021.174277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 11/24/2022]
Abstract
Cardiac hypertrophy (CH) plays a central role in cardiac remodeling and is an independent risk factor for cardiac events. It is imperative to find drugs with protective effect on CH. Dioscin, one natural product, shows various pharmacological activities, and PKCepsilon (PKCε) plays an important role in the physiological hypertrophic responses. Thus, we aimed to investigate the possible protective effect of dioscin on CH through PKCε. In the present study, the isoproterenol (ISO)-induced H9C2 cells and primary cardiomyocytes models, and the ISO-induced rat model were established, and the pharmacodynamics and mechanism of dioscin were investigated. In vitro results prompted that, dioscin significantly improved ISO-induced cardiomyocyte hypertrophy, decreased the levels of cell size, protein content of single cell, reactive oxygen species, atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), beta-myosin heavy chain (β-MHC). Moreover, in vivo, changes in histopathological of the animals caused by ISO are improved by dioscin. And dioscin decreased the index of CH and the levels of CK, MDA, LDH, and increased the levels of GSH, SOD and GSH-Px. Mechanism research showed that dioscin inhibited the expression levels of PKCε, and affected the expression levels of p-MEK, p-ERK, Nrf2, Keap1 and HO-1 to inhibit oxidative stress. In addition, the results of ISO-induced CH in PKCε siRNA transfected H9C2 cells and C57BL/6 mice further showed that the protective effect of dioscin on CH, which was mediated by inhibition of PKCε/ERK signal pathway. In summary, dioscin can effectively inhibit CH by regulating PKCε-mediated oxidative stress, which should be considered as one potent candidate for new drug research and development to treat CH in the future.
Collapse
Affiliation(s)
- Ruomiao Li
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Qianhui Yuan
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Meng Gao
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, China.
| | - Chuntong Liu
- Pharmaceutical Department, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, China.
| |
Collapse
|
32
|
Arangalage D, Degrauwe N, Michielin O, Monney P, Özdemir BC. Pathophysiology, diagnosis and management of cardiac toxicity induced by immune checkpoint inhibitors and BRAF and MEK inhibitors. Cancer Treat Rev 2021; 100:102282. [PMID: 34438238 DOI: 10.1016/j.ctrv.2021.102282] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/16/2022]
Abstract
Immune checkpoint inhibitors (ICIs) and BRAF and MEK inhibitors (BRAFi/MEKi) have drastically improved the outcome of melanoma patients. ICIs can induce myocarditis, a rare immune related adverse event (irAE) with an estimated lethality of 50%. BRAFi/MEKi may induce left ventricular ejection fraction decrease, hypertension or QT interval prolongation. While the BRAFi/MEKi induced cardiotoxicity is often reversible upon treatment discontinuation or dose adaptation and symptomatic therapy is often sufficient to restore cardiac function, the treatment of ICI-induced myocarditis mainly relies on high dose corticosteroids. There is no established therapy for steroid resistant myocarditis, yet various drugs have been reported to improve outcome. Shared epitopes between melanoma cells and cardiac tissue are thought to underlie the development of ICIs induced myocarditis. The mechanism of BRAFi/MEKi induced cardiotoxicity appears to be related to the Ras-Raf-MEK-ERK pathway in cardiomyocyte repair, survival and proliferation. With the emerging application of ICI-BRAFi/MEKi combinations, so called triplet therapies, differentiating between these two types of cardiotoxicity will become important for appropriate patient management. In this article we provide a summary of the existing literature on the pathophysiology, diagnosis and management of cardiotoxicity of melanoma therapies.
Collapse
Affiliation(s)
- Dimitri Arangalage
- Department of Cardiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Nils Degrauwe
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Olivier Michielin
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Pierre Monney
- Department of Cardiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland.
| | - Berna C Özdemir
- Department of Oncology, Bern University Hospital (Inselspital), University of Bern, Switzerland; International Cancer Prevention Institute, Epalinges, Switzerland.
| |
Collapse
|
33
|
Meijles DN, Cull JJ, Cooper ST, Markou T, Hardyman MA, Fuller SJ, Alharbi HO, Haines ZH, Alcantara-Alonso V, Glennon PE, Sheppard MN, Sugden PH, Clerk A. The anti-cancer drug dabrafenib is not cardiotoxic and inhibits cardiac remodelling and fibrosis in a murine model of hypertension. Clin Sci (Lond) 2021; 135:1631-1647. [PMID: 34296750 PMCID: PMC8302807 DOI: 10.1042/cs20210192] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/30/2022]
Abstract
Raf kinases signal via extracellular signal-regulated kinases 1/2 (ERK1/2) to drive cell division. Since activating mutations in BRAF (B-Raf proto-oncogene, serine/threonine kinase) are highly oncogenic, BRAF inhibitors including dabrafenib have been developed for cancer. Inhibitors of ERK1/2 signalling used for cancer are cardiotoxic in some patients, raising the question of whether dabrafenib is cardiotoxic. In the heart, ERK1/2 signalling promotes not only cardiomyocyte hypertrophy and is cardioprotective but also promotes fibrosis. Our hypothesis is that ERK1/2 signalling is not required in a non-stressed heart but is required for cardiac remodelling. Thus, dabrafenib may affect the heart in the context of, for example, hypertension. In experiments with cardiomyocytes, cardiac fibroblasts and perfused rat hearts, dabrafenib inhibited ERK1/2 signalling. We assessed the effects of dabrafenib (3 mg/kg/d) on male C57BL/6J mouse hearts in vivo. Dabrafenib alone had no overt effects on cardiac function/dimensions (assessed by echocardiography) or cardiac architecture. In mice treated with 0.8 mg/kg/d angiotensin II (AngII) to induce hypertension, dabrafenib inhibited ERK1/2 signalling and suppressed cardiac hypertrophy in both acute (up to 7 d) and chronic (28 d) settings, preserving ejection fraction. At the cellular level, dabrafenib inhibited AngII-induced cardiomyocyte hypertrophy, reduced expression of hypertrophic gene markers and almost completely eliminated the increase in cardiac fibrosis both in interstitial and perivascular regions. Dabrafenib is not overtly cardiotoxic. Moreover, it inhibits maladaptive hypertrophy resulting from AngII-induced hypertension. Thus, Raf is a potential therapeutic target for hypertensive heart disease and drugs such as dabrafenib, developed for cancer, may be used for this purpose.
Collapse
Affiliation(s)
- Daniel N. Meijles
- School of Biological Sciences, University of Reading, Reading RG6 2AS, U.K
- Molecular and Clinical Sciences Institute, St George’s University of London, London SW17 0RE, U.K
| | - Joshua J. Cull
- School of Biological Sciences, University of Reading, Reading RG6 2AS, U.K
| | - Susanna T.E. Cooper
- Molecular and Clinical Sciences Institute, St George’s University of London, London SW17 0RE, U.K
| | - Thomais Markou
- School of Biological Sciences, University of Reading, Reading RG6 2AS, U.K
| | | | - Stephen J. Fuller
- School of Biological Sciences, University of Reading, Reading RG6 2AS, U.K
| | - Hajed O. Alharbi
- School of Biological Sciences, University of Reading, Reading RG6 2AS, U.K
| | - Zoe H.R. Haines
- Molecular and Clinical Sciences Institute, St George’s University of London, London SW17 0RE, U.K
| | | | - Peter E. Glennon
- University Hospitals Coventry and Warwickshire, University Hospital Cardiology Department, Clifford Bridge Road, Coventry CV2 2DX, U.K
| | - Mary N. Sheppard
- Molecular and Clinical Sciences Institute, St George’s University of London, London SW17 0RE, U.K
- CRY Cardiovascular Pathology Department, St. George's University of London, London, U.K
- St. George’s Healthcare NHS Trust, London, U.K
| | - Peter H. Sugden
- School of Biological Sciences, University of Reading, Reading RG6 2AS, U.K
| | - Angela Clerk
- School of Biological Sciences, University of Reading, Reading RG6 2AS, U.K
| |
Collapse
|
34
|
Trametinib-associated Hyponatremia in a Child With Low-grade Glioma is Not Seen Following Treatment With Alternative MEK Inhibitor. J Pediatr Hematol Oncol 2021; 43:e550-e553. [PMID: 32520842 DOI: 10.1097/mph.0000000000001859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/05/2020] [Indexed: 01/22/2023]
Abstract
Molecularly targeted therapy with MEK inhibitors has been increasingly incorporated into the treatment of pediatric low-grade gliomas, but this promising therapy is associated with distinctive and specific toxicities. Understanding life-threatening MEK inhibitor toxicities and their management is critical to MEK inhibitor safety, especially among young children. This report describes severe hyponatremia associated with trametinib in an infant with progressive low-grade glioma without underlying endocrine dysfunction, which recurred despite significant dose reduction. Therapy with an alternative MEK inhibitor, binimetinib, provided excellent tumor response without hyponatremia, suggesting that some toxicities may be avoided by changing MEK inhibitor agents within the same class.
Collapse
|
35
|
Mudd TW, Khalid M, Guddati AK. Cardiotoxicity of chemotherapy and targeted agents. Am J Cancer Res 2021; 11:1132-1147. [PMID: 33948350 PMCID: PMC8085845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/04/2021] [Indexed: 06/12/2023] Open
Abstract
The evolution of cancer treatment and development of new classes of anticancer therapies have continued to revolutionize the field of oncology. New therapies including targeted agents, immunotherapies, and adoptive cell transfer have allowed for exciting survival benefit progress for patients. However, the novel nature of these therapies as well as the longer survival periods of patients receiving them has highlighted the various side effects of anticancer therapies. Cardiotoxicity has emerged as a major side effect of anticancer treatment and can present both acutely during treatment and chronically even years after treatment has been completed. This work compiles the cardiotoxic side effects of various chemotherapeutic and targeted anticancer therapies and their management.
Collapse
Affiliation(s)
- Todd William Mudd
- Division of Hematology/Oncology Georgia Cancer Center, Augusta UniversityAugusta 30909, GA, USA
| | | | - Achuta Kumar Guddati
- Division of Hematology/Oncology Georgia Cancer Center, Augusta UniversityAugusta 30909, GA, USA
| |
Collapse
|
36
|
Wang C, Wang H, Zheng C, Liu Z, Gao X, Xu F, Niu Y, Zhang L, Xu P. Research progress of MEK1/2 inhibitors and degraders in the treatment of cancer. Eur J Med Chem 2021; 218:113386. [PMID: 33774345 DOI: 10.1016/j.ejmech.2021.113386] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/25/2021] [Accepted: 03/13/2021] [Indexed: 12/14/2022]
Abstract
Mitogen-activated protein kinase kinases 1 and 2 (MEK1/2) are the crucial part of the RAS-RAF-MEK-ERK pathway (or ERK pathway), which is involved in the regulation of various cellular processes including proliferation, survival, and differentiation et al. Targeting MEK has become an important strategy for cancer therapy, and 4 MEK inhibitors (MEKis) have been approved by FDA to date. However, the application of MEKis is limited due to acquired resistance under long-term treatment. Fortunately, an emerging technology, named proteolysis targeting chimera (PROTAC), could break through this limitation by inducing MEK1/2 degradation. Compared to MEKis, MEK1/2 PROTAC is rarely studied and only three MEK1/2 PROTAC molecules, have been reported until now. This paper will outline the ERK pathway and the mechanism and research progress of MEK1/2 inhibitors, but focus on the development of MEK degraders and their optimization strategies. PAC-1 strategy which can induce MEK degradation indirectly, other PROTACs on ERK pathway, the advantages and challenges of PROTAC technology will be subsequently discussed.
Collapse
Affiliation(s)
- Chao Wang
- National Pharmaceutical Teaching Laboratory Center, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Han Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Cangxin Zheng
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Xiaozuo Gao
- Royal Melbourne Institute of Technology University, Melbourne, Australia
| | - Fengrong Xu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yan Niu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Ping Xu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| |
Collapse
|
37
|
Thomas K, Ayse C, Natalia K, Peter B, Bedriye SH, Praveen G, Hakan A, Markus S, Wolfgang S, Yeong-Hoon C, Miroslav B, Manfred R. The MEK/ERK Module Is Reprogrammed in Remodeling Adult Cardiomyocytes. Int J Mol Sci 2020; 21:ijms21176348. [PMID: 32882982 PMCID: PMC7503571 DOI: 10.3390/ijms21176348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/26/2020] [Accepted: 08/30/2020] [Indexed: 12/18/2022] Open
Abstract
Fetal and hypertrophic remodeling are hallmarks of cardiac restructuring leading chronically to heart failure. Since the Ras/Raf/MEK/ERK cascade (MAPK) is involved in the development of heart failure, we hypothesized, first, that fetal remodeling is different from hypertrophy and, second, that remodeling of the MAPK occurs. To test our hypothesis, we analyzed models of cultured adult rat cardiomyocytes as well as investigated myocytes in the failing human myocardium by western blot and confocal microscopy. Fetal remodeling was induced through endothelial morphogens and monitored by the reexpression of Acta2, Actn1, and Actb. Serum-induced hypertrophy was determined by increased surface size and protein content of cardiomyocytes. Serum and morphogens caused reprogramming of Ras/Raf/MEK/ERK. In both models H-Ras, N-Ras, Rap2, B- and C-Raf, MEK1/2 as well as ERK1/2 increased while K-Ras was downregulated. Atrophy, MAPK-dependent ischemic resistance, loss of A-Raf, and reexpression of Rap1 and Erk3 highlighted fetal remodeling, while A-Raf accumulation marked hypertrophy. The knock-down of B-Raf by siRNA reduced MAPK activation and fetal reprogramming. In conclusion, we demonstrate that fetal and hypertrophic remodeling are independent processes and involve reprogramming of the MAPK.
Collapse
Affiliation(s)
- Kubin Thomas
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, 61231 Bad Nauheim, Germany; (C.A.); (K.N.); (G.P.); (S.M.); (C.Y.-H.)
- Campus Kerckhoff, Justus-Liebig-University Giessen, 61231 Bad Nauheim, Germany
- Correspondence: (K.T.); (B.M.); (R.M.)
| | - Cetinkaya Ayse
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, 61231 Bad Nauheim, Germany; (C.A.); (K.N.); (G.P.); (S.M.); (C.Y.-H.)
- Campus Kerckhoff, Justus-Liebig-University Giessen, 61231 Bad Nauheim, Germany
| | - Kubin Natalia
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, 61231 Bad Nauheim, Germany; (C.A.); (K.N.); (G.P.); (S.M.); (C.Y.-H.)
- Campus Kerckhoff, Justus-Liebig-University Giessen, 61231 Bad Nauheim, Germany
| | - Bramlage Peter
- Institute for Pharmacology and Preventive Medicine, Bahnhofstraße 20, 49661 Cloppenburg, Germany;
| | - Sen-Hild Bedriye
- Pediatric Heart Center, Justus Liebig University, Feulgenstrasse 10-12, 35392 Giessen, Germany; (S.-H.B.); (A.H.)
| | - Gajawada Praveen
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, 61231 Bad Nauheim, Germany; (C.A.); (K.N.); (G.P.); (S.M.); (C.Y.-H.)
- Campus Kerckhoff, Justus-Liebig-University Giessen, 61231 Bad Nauheim, Germany
| | - Akintürk Hakan
- Pediatric Heart Center, Justus Liebig University, Feulgenstrasse 10-12, 35392 Giessen, Germany; (S.-H.B.); (A.H.)
| | - Schönburg Markus
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, 61231 Bad Nauheim, Germany; (C.A.); (K.N.); (G.P.); (S.M.); (C.Y.-H.)
- Campus Kerckhoff, Justus-Liebig-University Giessen, 61231 Bad Nauheim, Germany
| | - Schaper Wolfgang
- Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany;
| | - Choi Yeong-Hoon
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, 61231 Bad Nauheim, Germany; (C.A.); (K.N.); (G.P.); (S.M.); (C.Y.-H.)
- Campus Kerckhoff, Justus-Liebig-University Giessen, 61231 Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site RhineMain, 60590 Frankfurt/Main, Germany
| | - Barancik Miroslav
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
- Correspondence: (K.T.); (B.M.); (R.M.)
| | - Richter Manfred
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, 61231 Bad Nauheim, Germany; (C.A.); (K.N.); (G.P.); (S.M.); (C.Y.-H.)
- Campus Kerckhoff, Justus-Liebig-University Giessen, 61231 Bad Nauheim, Germany
- Correspondence: (K.T.); (B.M.); (R.M.)
| |
Collapse
|
38
|
Klesse LJ, Jordan JT, Radtke HB, Rosser T, Schorry E, Ullrich N, Viskochil D, Knight P, Plotkin SR, Yohay K. The Use of MEK Inhibitors in Neurofibromatosis Type 1-Associated Tumors and Management of Toxicities. Oncologist 2020; 25:e1109-e1116. [PMID: 32272491 PMCID: PMC7356675 DOI: 10.1634/theoncologist.2020-0069] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/05/2020] [Indexed: 12/31/2022] Open
Abstract
Early-phase clinical trials using oral inhibitors of MEK, the mitogen-activated protein kinase kinase, have demonstrated benefit for patients with neurofibromatosis type 1 (NF1)-associated tumors, particularly progressive low-grade gliomas and plexiform neurofibromas. Given this potential of MEK inhibition as an effective medical therapy, the use of targeted agents in the NF1 population is likely to increase substantially. For clinicians with limited experience prescribing MEK inhibitors, concern about managing these treatments may be a barrier to use. In this manuscript, the Clinical Care Advisory Board of the Children's Tumor Foundation reviews the published experience with MEK inhibitors in NF1 and outlines recommendations for side-effect management, as well as monitoring guidelines. These recommendations can serve as a beginning framework for NF providers seeking to provide the most effective treatments for their patients. IMPLICATIONS FOR PRACTICE: Neurofibromatosis type 1 (NF1) clinical care is on the cusp of a transformative shift. With the success of recent clinical trials using MEK inhibitors, an increasing number of NF1 patients are being treated with MEK inhibitors for both plexiform neurofibromas and low-grade gliomas. The use of MEK inhibitors is likely to increase substantially in NF1. Given these changes, the Clinical Care Advisory Board of the Children's Tumor Foundation has identified a need within the NF1 clinical community for guidance for the safe and effective use of MEK inhibitors for NF1-related tumors. This article provides a review of the published experience of MEK inhibitors in NF1 and provides recommendations for monitoring and management of side effects.
Collapse
Affiliation(s)
| | | | - Heather B. Radtke
- Medical College of WisconsinMilwaukeeWisconsinUSA
- Children's Tumor FoundationNew YorkNew YorkUSA
| | - Tena Rosser
- Keck School of Medicine of USC, Children's Hospital of Los AngelesLos AngelesCaliforniaUSA
| | - Elizabeth Schorry
- Cincinnati Children's Hospital, University of Cincinnati, CincinnatiOhio
| | - Nicole Ullrich
- Boston Children's Hospital, Dana Farber Cancer InstituteBostonMassachusettsUSA
| | | | | | | | | |
Collapse
|
39
|
Buckley AM, Lynam-Lennon N, O'Neill H, O'Sullivan J. Targeting hallmarks of cancer to enhance radiosensitivity in gastrointestinal cancers. Nat Rev Gastroenterol Hepatol 2020; 17:298-313. [PMID: 32005946 DOI: 10.1038/s41575-019-0247-2] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/26/2019] [Indexed: 12/19/2022]
Abstract
Radiotherapy is used in the treatment of approximately 50% of all malignancies including gastrointestinal cancers. Radiation can be given prior to surgery (neoadjuvant radiotherapy) to shrink the tumour or after surgery to kill any remaining cancer cells. Radiotherapy aims to maximize damage to cancer cells, while minimizing damage to healthy cells. However, only 10-30% of patients with rectal cancer or oesophageal cancer have a pathological complete response to neoadjuvant chemoradiation therapy, with the rest suffering the negative consequences of toxicities and delays to surgery with no clinical benefit. Furthermore, in pancreatic cancer, neoadjuvant chemoradiation therapy results in a pathological complete response in only 4% of patients and a partial pathological response in only 31%. Resistance to radiation therapy is polymodal and associated with a number of biological alterations both within the tumour itself and in the surrounding microenvironment including the following: altered cell cycle; repopulation by cancer stem cells; hypoxia; altered management of oxidative stress; evasion of apoptosis; altered DNA damage response and enhanced DNA repair; inflammation; and altered mitochondrial function and cellular energetics. Radiosensitizers are needed to improve treatment response to radiation, which will directly influence patient outcomes in gastrointestinal cancers. This article reviews the literature to identify strategies - including DNA-targeting agents, antimetabolic agents, antiangiogenics and novel immunotherapies - being used to enhance radiosensitivity in gastrointestinal cancers according to the hallmarks of cancer. Evidence from radiosensitizers from in vitro and in vivo models is documented and the action of radiosensitizers through clinical trial data is assessed.
Collapse
Affiliation(s)
- Amy M Buckley
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Hazel O'Neill
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Jacintha O'Sullivan
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
40
|
Tomasovic A, Brand T, Schanbacher C, Kramer S, Hümmert MW, Godoy P, Schmidt-Heck W, Nordbeck P, Ludwig J, Homann S, Wiegering A, Shaykhutdinov T, Kratz C, Knüchel R, Müller-Hermelink HK, Rosenwald A, Frey N, Eichler J, Dobrev D, El-Armouche A, Hengstler JG, Müller OJ, Hinrichs K, Cuello F, Zernecke A, Lorenz K. Interference with ERK-dimerization at the nucleocytosolic interface targets pathological ERK1/2 signaling without cardiotoxic side-effects. Nat Commun 2020; 11:1733. [PMID: 32265441 PMCID: PMC7138859 DOI: 10.1038/s41467-020-15505-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 03/13/2020] [Indexed: 12/16/2022] Open
Abstract
Dysregulation of extracellular signal-regulated kinases (ERK1/2) is linked to several diseases including heart failure, genetic syndromes and cancer. Inhibition of ERK1/2, however, can cause severe cardiac side-effects, precluding its wide therapeutic application. ERKT188-autophosphorylation was identified to cause pathological cardiac hypertrophy. Here we report that interference with ERK-dimerization, a prerequisite for ERKT188-phosphorylation, minimizes cardiac hypertrophy without inducing cardiac adverse effects: an ERK-dimerization inhibitory peptide (EDI) prevents ERKT188-phosphorylation, nuclear ERK1/2-signaling and cardiomyocyte hypertrophy, protecting from pressure-overload-induced heart failure in mice whilst preserving ERK1/2-activity and cytosolic survival signaling. We also examine this alternative ERK1/2-targeting strategy in cancer: indeed, ERKT188-phosphorylation is strongly upregulated in cancer and EDI efficiently suppresses cancer cell proliferation without causing cardiotoxicity. This powerful cardio-safe strategy of interfering with ERK-dimerization thus combats pathological ERK1/2-signaling in heart and cancer, and may potentially expand therapeutic options for ERK1/2-related diseases, such as heart failure and genetic syndromes. Drugs targeting dysregulated ERK1/2 signaling can cause severe cardiac side effects, precluding their wide therapeutic application. Here, a new and cardio-safe targeting strategy is presented that interferes with ERK dimerization to prevent pathological ERK1/2 signaling in the heart and cancer.
Collapse
Affiliation(s)
- Angela Tomasovic
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078, Würzburg, Germany.,Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., 44139, Dortmund, Germany
| | - Theresa Brand
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078, Würzburg, Germany.,Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., 44139, Dortmund, Germany
| | - Constanze Schanbacher
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078, Würzburg, Germany.,Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., 44139, Dortmund, Germany
| | - Sofia Kramer
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078, Würzburg, Germany
| | - Martin W Hümmert
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078, Würzburg, Germany.,Department of Neurology, Hannover Medical School, 30625, Hannover, Germany
| | - Patricio Godoy
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, 44139, Dortmund, Germany
| | - Wolfgang Schmidt-Heck
- Leibniz Institute for Natural Product Research and Infection Biology -Hans Knoell Institute-, 07745, Jena, Germany
| | - Peter Nordbeck
- Comprehensive Heart Failure Center, 97078, Würzburg, Germany
| | - Jonas Ludwig
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Susanne Homann
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078, Würzburg, Germany
| | - Armin Wiegering
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Timur Shaykhutdinov
- Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., 12489, Berlin, Germany
| | - Christoph Kratz
- Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., 12489, Berlin, Germany
| | - Ruth Knüchel
- Institute of Pathology, University Hospital Aachen, RWTH Aachen, 52074, Aachen, Germany
| | | | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, 97080, Würzburg, Germany
| | - Norbert Frey
- Department of Internal Medicine III, University of Kiel, 24105, Kiel, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Jutta Eichler
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, 45147, Essen, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, TU Dresden, 01307, Dresden, Germany
| | - Jan G Hengstler
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, 44139, Dortmund, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, 24105, Kiel, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Karsten Hinrichs
- Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., 12489, Berlin, Germany
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, University of Würzburg, 97080, Würzburg, Germany
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078, Würzburg, Germany. .,Leibniz-Institut für Analytische Wissenschaften - ISAS-e.V., 44139, Dortmund, Germany. .,Comprehensive Heart Failure Center, 97078, Würzburg, Germany.
| |
Collapse
|
41
|
Berger M, Amini-Adlé M, Maucort-Boulch D, Robinson P, Thomas L, Dalle S, Courand PY. Left ventricular ejection fraction decrease related to BRAF and/or MEK inhibitors in metastatic melanoma patients: A retrospective analysis. Cancer Med 2020; 9:2611-2620. [PMID: 32056395 PMCID: PMC7163110 DOI: 10.1002/cam4.2922] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 12/12/2022] Open
Abstract
BRAF and MEKis have revolutionized the management of BRAFV600‐mutated melanoma patients. Left ventricular ejection fraction decrease (LVEF‐D) related to these treatments has not been thoroughly evaluated to date. The main objective of this study was to describe characteristics of LVEF‐D in melanoma patients treated with BRAF and/or MEKis. Metastatic melanoma patients treated with BRAF and/or MEKis between March 1, 2012 and May 18, 2018 were included retrospectively (Lyon Sud University Hospital, Hospices Civils de Lyon). LVEF‐D was defined as a reduction in LVEF ≥10% from baseline to a value <55%; normalization was defined as a value ≥55%. Among the 88 patients included, 12 (13.6%) experienced a LVEF‐D, including 10 grade 2 and 2 grade 3. The median onset of which was 11 months (IQR [3‐21]). No patient previously treated with beta‐blockers (n = 12) experienced a LVEF‐D. Analysis of laboratory parameters, electrocardiogram, and transthoracic echocardiography during the follow‐up did not find any predictive marker of LVEF‐D. All patients who benefited from a specific treatment of LVEF‐D had a normalization of LVEF at the end of follow‐up. LVEF recovery was significantly better for patients treated with angiotensin converting enzyme inhibitors and beta‐blockers than those who did not (P = .019). Ophthalmological adverse events were significantly more frequent in patients who experienced a LVEF‐D (P = .006) and the latter did not influence overall‐survival (P = .117) or progression‐free‐survival (P = .297). LVEF‐D is a common and easily manageable adverse event due to BRAF and MEKis. Its association with ocular toxicity suggests a close ophthalmological monitoring when LVEF‐D occurs.
Collapse
Affiliation(s)
- Mathilde Berger
- Service de dermatologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Mona Amini-Adlé
- Service de dermatologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Delphine Maucort-Boulch
- Service de Biostatistiques, Hospices Civils de Lyon, Université Lyon 1, Lyon, France.,Laboratoire de Biométrie et Biologie Evolutive, Université de Lyon, CNRS, Equipe Biostatistique-Santé, Villeurbanne, France
| | - Philip Robinson
- Direction de la Recherche Clinique et de l'Innovation, Hospices Civils de Lyon, Lyon, France
| | - Luc Thomas
- Service de dermatologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France.,Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Université Claude Bernard, Lyon, France
| | - Stéphane Dalle
- Service de dermatologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France.,Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Université Claude Bernard, Lyon, France
| | - Pierre-Yves Courand
- Hospices Civils de Lyon, Service de cardiologie, Hôpital de la Croix-Rousse et Centre Hospitalier Lyon Sud, Lyon, France.,Université de Lyon, Creatis Umr Inserm U1044, INSA, Lyon, France
| |
Collapse
|
42
|
Lenihan DJ, Fradley MG, Dent S, Brezden-Masley C, Carver J, Filho RK, Neilan TG, Blaes A, Melloni C, Herrmann J, Armenian S, Thavendiranathan P, Armstrong GT, Ky B, Hajjar L. Proceedings From the Global Cardio-Oncology Summit: The Top 10 Priorities to Actualize for CardioOncology. JACC CardioOncol 2019; 1:256-272. [PMID: 34396188 PMCID: PMC8352295 DOI: 10.1016/j.jaccao.2019.11.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 11/04/2019] [Indexed: 12/27/2022] Open
Abstract
The discipline of cardio-oncology has expanded at a remarkable pace. Recent developments and challenges to clinicians who practice cardio-oncology were presented at the Global Cardio-Oncology Summit on October 3 to 4, 2019, in São Paulo, Brazil. Here, we present the top 10 priorities for our field that were discussed at the meeting, and also detail a potential path forward to address these challenges. Defining robust predictors of cardiotoxicity, clarifying the role of cardioprotection, managing and preventing thromboembolism, improving hematopoietic stem cell transplant outcomes, personalizing cardiac interventions, building the cardio-oncology community, detecting and treating cardiovascular events associated with immunotherapy, understanding tyrosine kinase inhibitor cardiotoxicity, and enhancing survivorship care are all priorities for the field. The path forward requires a commitment to research, education, and excellence in clinical care to improve our patients' lives.
Collapse
Key Words
- CV, cardiovascular
- CVD, cardiovascular disease
- DOAC, direct oral anticoagulant
- GCOS, Global Cardio-Oncology Summit
- GLS, global longitudinal strain
- HCT, hematopoietic cell transplantation
- ICI, immune checkpoint inhibitor
- LVEF, left ventricular ejection fraction
- PD-1, programmed cell death 1 or its ligand
- PD-L1, programmed cell death ligand 1
- TKI, tyrosine kinase inhibitor
- VTE, venous thromboembolism
- anthracycline
- antiangiogenic therapy
- bone marrow transplantation
- breast cancer
- cancer survivorship
- immunotherapy
- thrombosis
- tyrosine kinase inhibitor
Collapse
Affiliation(s)
- Daniel J. Lenihan
- Cardio-Oncology Center of Excellence, Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Michael G. Fradley
- Cardio-Oncology Program, Moffitt Cancer Center, University of South Florida, Tampa, Florida, USA
| | - Susan Dent
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Joseph Carver
- Cardio-Oncology Center of Excellence at the Abramson Cancer Center, Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Roberto Kalil Filho
- Cardio-Oncology Program, Department of Cardiopneumology, Cancer Institute and Heart Institute, University of São Paulo, Brazil
| | - Tomas G. Neilan
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anne Blaes
- Division of Hematology/Oncology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Chiara Melloni
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Saro Armenian
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Paaladinesh Thavendiranathan
- Division of Cardiology, Peter Munk Cardiac Centre, Ted Rogers Program in Cardiotoxicity Prevention, University Health Network, University of Toronto, Toronto, Canada
| | - Gregory T. Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Bonnie Ky
- Cardio-Oncology Center of Excellence at the Abramson Cancer Center, Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ludhmila Hajjar
- Cardio-Oncology Program, Department of Cardiopneumology, Cancer Institute and Heart Institute, University of São Paulo, Brazil
| |
Collapse
|
43
|
[Supportive therapy and management of side effects in dermato-oncology]. Hautarzt 2019; 70:975-988. [PMID: 31720719 DOI: 10.1007/s00105-019-04496-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In the context of supportive therapy, possible complaints which may be caused by the cancer itself, by the antitumoral therapy or by psychosocial concerns are considered. Due to the introduction of new anticancer drugs in dermato-oncology, clinicians are confronted with a novel spectrum of adverse events. There are a number of inflammatory, immune-mediated side effects caused by immunotherapies, which can affect virtually any organ. Targeted therapies also have specific side effects. Basically, the management of adverse events depends on their severity. Besides treatment breaks and dosage modifications, immunotherapy-related adverse events are treated with systemic immunosuppressants. Supportive symptomatic therapy is offered. The additional consideration of psychosocial problems can improve quality of life of cancer patients.
Collapse
|
44
|
Matsui T, Miyamoto K, Yamanaka K, Okai Y, Kaushik EP, Harada K, Wagoner M, Shinozawa T. Cell-based two-dimensional morphological assessment system to predict cancer drug-induced cardiotoxicity using human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Appl Pharmacol 2019; 383:114761. [DOI: 10.1016/j.taap.2019.114761] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/31/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022]
|
45
|
Guo Y, Sui JY, Kim K, Zhang Z, Qu XA, Nam YJ, Willette RN, Barnett JV, Knollmann BC, Force T, Lal H. Cardiomyocyte Homeodomain-Interacting Protein Kinase 2 Maintains Basal Cardiac Function via Extracellular Signal-Regulated Kinase Signaling. Circulation 2019; 140:1820-1833. [PMID: 31581792 DOI: 10.1161/circulationaha.119.040740] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cardiac kinases play a critical role in the development of heart failure, and represent potential tractable therapeutic targets. However, only a very small fraction of the cardiac kinome has been investigated. To identify novel cardiac kinases involved in heart failure, we used an integrated transcriptomics and bioinformatics analysis and identified Homeodomain-Interacting Protein Kinase 2 (HIPK2) as a novel candidate kinase. The role of HIPK2 in cardiac biology is unknown. METHODS We used the Expression2Kinase algorithm for the screening of kinase targets. To determine the role of HIPK2 in the heart, we generated cardiomyocyte (CM)-specific HIPK2 knockout and heterozygous mice. Heart function was examined by echocardiography, and related cellular and molecular mechanisms were examined. Adeno-associated virus serotype 9 carrying cardiac-specific constitutively active MEK1 (TnT-MEK1-CA) was administrated to rescue cardiac dysfunction in CM-HIPK2 knockout mice. RESULTS To our knowledge, this is the first study to define the role of HIPK2 in cardiac biology. Using multiple HIPK2 loss-of-function mouse models, we demonstrated that reduction of HIPK2 in CMs leads to cardiac dysfunction, suggesting a causal role in heart failure. It is important to note that cardiac dysfunction in HIPK2 knockout mice developed with advancing age, but not during development. In addition, CM-HIPK2 knockout mice and CM-HIPK2 heterozygous mice exhibited a gene dose-response relationship of CM-HIPK2 on heart function. HIPK2 expression in the heart was significantly reduced in human end-stage ischemic cardiomyopathy in comparison to nonfailing myocardium, suggesting a clinical relevance of HIPK2 in cardiac biology. In vitro studies with neonatal rat ventricular CMscorroborated the in vivo findings. Specifically, adenovirus-mediated overexpression of HIPK2 suppressed the expression of heart failure markers, NPPA and NPPB, at basal condition and abolished phenylephrine-induced pathological gene expression. An array of mechanistic studies revealed impaired extracellular signal-regulated kinase 1/2 signaling in HIPK2-deficient hearts. An in vivo rescue experiment with adeno-associated virus serotype 9 TnT-MEK1-CA nearly abolished the detrimental phenotype of knockout mice, suggesting that impaired extracellular signal-regulated kinase signaling mediated apoptosis as the key factor driving the detrimental phenotype in CM-HIPK2 knockout mice hearts. CONCLUSIONS Taken together, these findings suggest that CM-HIPK2 is required to maintain normal cardiac function via extracellular signal-regulated kinase signaling.
Collapse
Affiliation(s)
- Yuanjun Guo
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (Y. Guo, J.V.B.)
| | - Jennifer Y Sui
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN
| | - Kyungsoo Kim
- Division of Clinical Pharmacology (K.K., B.C.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Zhentao Zhang
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN.,Department of Cell and Developmental Biology (Z.Z., Y.-J.N.), Vanderbilt University, Nashville, TN.,Vanderbilt Center for Stem Cell Biology (Z.Z., Y.-J.N.), Vanderbilt University, Nashville, TN
| | - Xiaoyan A Qu
- PAREXEL International, Research Triangle Park, Durham, NC (X.A.Q.)
| | - Young-Jae Nam
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN.,Department of Cell and Developmental Biology (Z.Z., Y.-J.N.), Vanderbilt University, Nashville, TN.,Vanderbilt Center for Stem Cell Biology (Z.Z., Y.-J.N.), Vanderbilt University, Nashville, TN
| | - Robert N Willette
- Heart Failure Discovery Performance Unit, Metabolic Pathways and Cardiovascular Therapeutic Area GlaxoSmithKline, King of Prussia, PA (R.N.W.)
| | - Joey V Barnett
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN (Y. Guo, J.V.B.)
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology (K.K., B.C.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Thomas Force
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN
| | - Hind Lal
- Division of Cardiovascular Medicine (Y.G., J.Y.S., Z.Z., Y.-J.N., T.F., H.L.), Vanderbilt University Medical Center, Nashville, TN.,Division of Cardiovascular Disease, University of Alabama at Birmingham, AL (H.L.)
| |
Collapse
|
46
|
Gogas HJ, Flaherty KT, Dummer R, Ascierto PA, Arance A, Mandala M, Liszkay G, Garbe C, Schadendorf D, Krajsova I, Gutzmer R, Sileni VC, Dutriaux C, de Groot JWB, Yamazaki N, Loquai C, Gollerkeri A, Pickard MD, Robert C. Adverse events associated with encorafenib plus binimetinib in the COLUMBUS study: incidence, course and management. Eur J Cancer 2019; 119:97-106. [PMID: 31437754 DOI: 10.1016/j.ejca.2019.07.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Dual inhibition of the mitogen-activated protein kinase pathway with BRAF/MEK inhibitor (BRAFi/MEKi) therapy is a standard treatment for BRAFV600-mutant metastatic melanoma and has historically been associated with grade III pyrexia or photosensitivity depending on the combination used. The objective of this study was to fully describe adverse events from the COLUMBUS study evaluating the most recent BRAF/MEK inhibitor combination encorafenib+binimetinib. PATIENTS AND METHODS Patients with locally advanced, unresectable or metastatic BRAFV600-mutant melanoma were randomised to receive encorafenib 450 mg once daily plus binimetinib 45 mg twice daily, encorafenib 300 mg once daily or vemurafenib 960 mg twice daily. Adverse events that represent known effects of available BRAFi and/or MEKi were evaluated. RESULTS The safety population included a total of 570 patients (encorafenib+binimetinib = 192; encorafenib = 192; vemurafenib = 186). Median duration of exposure was longer with encorafenib+binimetinib (51 weeks) than with encorafenib (31 weeks) or vemurafenib (27 weeks). Common BRAFi/MEKi toxicities with encorafenib+binimetinib were generally manageable, reversible and infrequently associated with discontinuation. Pyrexia was less frequent with encorafenib+binimetinib (18%) and encorafenib (16%) than with vemurafenib (30%) and occurred later in the course of therapy with encorafenib+binimetinib (median time to first onset: 85 days versus 2.5 days and 19 days, respectively). The incidence of photosensitivity was lower with encorafenib+binimetinib (5%) and encorafenib (4%) than with vemurafenib (30%). The incidence of serous retinopathy was higher with encorafenib+binimetinib (20%) than with encorafenib (2%) or vemurafenib (2%), but no patients discontinued encorafenib+binimetinib because of this event. CONCLUSION Encorafenib+binimetinib is generally well tolerated and has a low discontinuation rate in patients with BRAFV600-mutant melanoma, with a distinct safety profile as compared with other anti-BRAF/MEK targeted therapies. TRIAL REGISTRATION ClinicalTrials.gov (Identifier: NCT01909453) and with EudraCT (number 2013-001176-38).
Collapse
Affiliation(s)
- Helen J Gogas
- Department of Internal Medicine, National and Kapodistrian University of Athens, Laikon Hospital, Athens, Greece.
| | | | - Reinhard Dummer
- Department of Dermatology, University Hospital Zürich Skin Cancer Center and University Zürich, Zürich, Switzerland
| | - Paolo A Ascierto
- Melanoma Unit, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Ana Arance
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Mario Mandala
- Department of Oncology and Haematology, Papa Giovanni XXIII Cancer Center Hospital, Bergamo, Italy
| | - Gabriella Liszkay
- Department of Dermatology, National Institute of Oncology, Budapest, Hungary
| | - Claus Garbe
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany
| | - Ivana Krajsova
- Department of Dermato-oncology, University Hospital Prague and Charles University First Medical Faculty, Prague, Czech Republic
| | - Ralf Gutzmer
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| | | | - Caroline Dutriaux
- Department of Oncologic Dermatology, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux, France
| | | | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Carmen Loquai
- Department of Dermatology, University Medical Center Mainz, Mainz, Germany
| | | | | | - Caroline Robert
- Service of Dermatology, Department of Medicine and Paris-Sud University, Gustave Roussy, Villejuif, France
| |
Collapse
|
47
|
Gad MM, Saad AM, Al-Husseini MJ, Abushouk AI, Salahia S, Rehman KA, Riaz H, Ahmed HM. Temporal trends, ethnic determinants, and short-term and long-term risk of cardiac death in cancer patients: a cohort study. Cardiovasc Pathol 2019; 43:107147. [PMID: 31494524 DOI: 10.1016/j.carpath.2019.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/14/2019] [Accepted: 08/02/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND We evaluated the risk of cardiac death in patients with prior cancer diagnoses and compared risk by cancer type and ethnicity in a large US population. METHOD Utilizing the Surveillance, Epidemiology, and End Results database, data on patients with a cancer diagnosis between 2000 and 2014 were obtained. We calculated the standardized mortality ratio (SMR) of cardiac death after a cancer diagnosis and the excess risk per 10,000 person-years. We stratified the analysis according to the time interval between cancer and cardiac events, cancer site, cancer stage, and race. RESULTS A total of 4,671,989 patients with a cancer diagnosis were included, of which 163,255 died due to cardiac causes within 10 years of diagnosis. We found a significantly higher rate of cardiac death for cancer patients [SMR=1.16, 95% confidence interval (CI) 1.15-1.16] compared to the general population. When observed for each cancer site, the highest SMR was after a diagnosis of hepatocellular carcinoma (SMR=2.58, 95% CI 2.45-2.72), pancreatic cancer (SMR=2.36, 95% CI 2.25-2.47), and lung cancer (SMR=2.30, 95% CI 2.27-2.34). Patients with metastatic disease had a higher rate of cardiac death (SMR=2.16, 95% CI 2.13-2.19). When stratified by ethnicity, SMR for cardiac death was 1.76, 2.28, 3.68, 2.65, and 1.84 for whites, blacks, American Indians/Alaska Natives, Asians/Pacific Islanders, and Hispanics, respectively. CONCLUSIONS Cancer patients are more vulnerable to cardiac death than the general population, especially those with nonwhite ethnicity; liver, lung, and pancreatic cancers; and history of metastasis. Healthcare providers should be aware of this risk and pay particular attention to the highest-risk groups.
Collapse
Affiliation(s)
- Mohamed M Gad
- Cleveland Clinic, OH, USA; Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Anas M Saad
- Cleveland Clinic, OH, USA; Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Sami Salahia
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | | |
Collapse
|
48
|
Mincu RI, Mahabadi AA, Michel L, Mrotzek SM, Schadendorf D, Rassaf T, Totzeck M. Cardiovascular Adverse Events Associated With BRAF and MEK Inhibitors: A Systematic Review and Meta-analysis. JAMA Netw Open 2019; 2:e198890. [PMID: 31397860 PMCID: PMC6692687 DOI: 10.1001/jamanetworkopen.2019.8890] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
IMPORTANCE Cardiovascular adverse events (CVAEs) after treatment with BRAF and MEK inhibitors in patients with melanoma remain incompletely characterized. OBJECTIVE To determine the association of BRAF and MEK inhibitor treatment with CVAEs in patients with melanoma compared with BRAF inhibitor monotherapy. DATA SOURCES PubMed, Cochrane, and Web of Science were systematically searched for keywords vemurafenib, dabrafenib, encorafenib, trametinib, binimetinib, and cobinimetinib from database inception through November 30, 2018. STUDY SELECTION Randomized clinical trials reporting on CVAEs in patients with melanoma being treated with BRAF and MEK inhibitors compared with patients with melanoma being treated with BRAF inhibitor monotherapy were selected. DATA EXTRACTION AND SYNTHESIS Data assessment followed the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. Pooled relative risks (RRs) and 95% CIs were determined using random-effects and fixed-effects analyses. Subgroup analyses were conducted to assess study-level characteristics associated with CVAEs. MAIN OUTCOMES AND MEASURES The selected end points were pulmonary embolism, a decrease in left ventricular ejection fraction, arterial hypertension, myocardial infarction, atrial fibrillation, and QTc interval prolongation. All-grade and high-grade (≥3) CVAEs were recorded. RESULTS Overall, 5 randomized clinical trials including 2317 patients with melanoma were selected. Treatment with BRAF and MEK inhibitors was associated with an increased risk of pulmonary embolism (RR, 4.36; 95% CI, 1.23-15.44; P = .02), a decrease in left ventricular ejection fraction (RR, 3.72; 95% CI, 1.74-7.94; P < .001), and arterial hypertension (RR, 1.49; 95% CI, 1.12-1.97; P = .005) compared with BRAF inhibitor monotherapy. The RRs for myocardial infarction, atrial fibrillation, and QTc prolongation were similar between the groups. These results were consistent when assessing high-grade CVAEs (left ventricular ejection fraction: RR, 2.79; 95% CI, 1.36-5.73; P = .005; I2 = 29%; high-grade arterial hypertension: RR, 1.54; 95% CI, 1.14-2.08; P = .005; I2 = 0%), but RRs for high-grade pulmonary embolism were similar between groups. A higher risk of a decrease in left ventricular ejection fraction was associated with patients with a mean age younger than 55 years (RR, 26.50; 95% CI, 3.58-196.10; P = .001), and the associated risk of pulmonary embolism was higher for patients with a mean follow-up time longer than 15 months (RR, 7.70; 95% CI, 1.40-42.12; P = .02). CONCLUSIONS AND RELEVANCE Therapy with BRAF and MEK inhibitors was associated with a higher risk of CVAEs compared with BRAF inhibitor monotherapy. The findings may help to balance between beneficial melanoma treatment and cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Raluca I. Mincu
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Essen, Germany
| | - Amir A. Mahabadi
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Essen, Germany
| | - Lars Michel
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Essen, Germany
| | - Simone M. Mrotzek
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Essen, Germany
| | - Dirk Schadendorf
- West German Cancer Center, Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Tienush Rassaf
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Essen, Germany
| | - Matthias Totzeck
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
49
|
Ricard N, Scott RP, Booth CJ, Velazquez H, Cilfone NA, Baylon JL, Gulcher JR, Quaggin SE, Chittenden TW, Simons M. Endothelial ERK1/2 signaling maintains integrity of the quiescent endothelium. J Exp Med 2019; 216:1874-1890. [PMID: 31196980 PMCID: PMC6683988 DOI: 10.1084/jem.20182151] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/12/2019] [Accepted: 05/08/2019] [Indexed: 12/22/2022] Open
Abstract
The endothelial ERK1/2 pathway plays a crucial role in the maintenance of endothelial homeostasis. Its suppression results in activation of TGFβ signaling, leading to hypertension, renal failure, fibrosis, and sudden death, findings similar to those observed in patients treated with anti-VEGF agents. To define the role of ERK1/2 signaling in the quiescent endothelium, we induced endothelial Erk2 knockout in adult Erk1−/− mice. This resulted in a rapid onset of hypertension, a decrease in eNOS expression, and an increase in endothelin-1 plasma levels, with all mice dying within 5 wk. Immunostaining and endothelial fate mapping showed a robust increase in TGFβ signaling leading to widespread endothelial-to-mesenchymal transition (EndMT). Fibrosis affecting the cardiac conduction system was responsible for the universal lethality in these mice. Other findings included renal endotheliosis, loss of fenestrated endothelia in endocrine organs, and hemorrhages. An ensemble computational intelligence strategy, comprising deep learning and probabilistic programing of RNA-seq data, causally linked the loss of ERK1/2 in HUVECs in vitro to activation of TGFβ signaling, EndMT, suppression of eNOS, and induction of endothelin-1 expression. All in silico predictions were verified in vitro and in vivo. In summary, these data establish the key role played by ERK1/2 signaling in the maintenance of vascular normalcy.
Collapse
Affiliation(s)
- Nicolas Ricard
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Rizaldy P Scott
- Feinberg Cardiovascular and Renal Research Institute, Division of Nephrology/Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Carmen J Booth
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT
| | - Heino Velazquez
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, Yale University, New Haven, CT
| | - Nicholas A Cilfone
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE, Cambridge, MA.,Complex Biological Systems Alliance, Medford, MA
| | - Javier L Baylon
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE, Cambridge, MA.,Complex Biological Systems Alliance, Medford, MA
| | | | - Susan E Quaggin
- Feinberg Cardiovascular and Renal Research Institute, Division of Nephrology/Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Thomas W Chittenden
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE, Cambridge, MA .,Complex Biological Systems Alliance, Medford, MA.,Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT .,Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
50
|
Heinzerling L, Eigentler TK, Fluck M, Hassel JC, Heller-Schenck D, Leipe J, Pauschinger M, Vogel A, Zimmer L, Gutzmer R. Tolerability of BRAF/MEK inhibitor combinations: adverse event evaluation and management. ESMO Open 2019; 4:e000491. [PMID: 31231568 PMCID: PMC6555610 DOI: 10.1136/esmoopen-2019-000491] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/06/2019] [Accepted: 03/31/2019] [Indexed: 12/11/2022] Open
Abstract
The inhibition of the mitogen-activated protein kinases signalling pathway through combined use of BRAF and MEK inhibitors (BRAFi+MEKi) represents an established therapeutic option in patients with BRAF-mutated, advanced melanoma. These efficient therapies are well tolerated with mostly moderate and reversible side effects and a discontinuation rate due to adverse events of 11.5%-15.7%. Median duration of therapy ranges between 8.8 and 11.7 months. Based on data from confirmatory trials, safety profiles of three BRAFi+MEKi combinations were reviewed, that is, dabrafenib plus trametinib, vemurafenib plus cobimetinib and encorafenib plus binimetinib. Many adverse events are class effects, such as cutaneous, gastrointestinal, ocular, cardiac and musculoskeletal events; some adverse events are substance associated. Fever (dabrafenib) and photosensitivity (vemurafenib) are the most common and clinically prominent examples. Other adverse events are less frequent and the association to one substance is less strong such as anaemia, facial paresis (encorafenib), neutropenia (dabrafenib), skin rash, QTc prolongation and increased liver function tests (vemurafenib). This narrative review provides recommendations for monitoring, adverse event evaluation and management focusing on the clinically relevant side effects of the three regimens.
Collapse
Affiliation(s)
- Lucie Heinzerling
- Department of Dermatology, University of Erlangen, Erlangen, Germany
| | - Thomas K Eigentler
- Department of Dermatology, Center for Dermatooncology, University Medical Center Tübingen, Tübingen, Germany
| | - Michael Fluck
- Department of Internal Medicine, Fachklinik Hornheide, Münster, Germany
| | - Jessica C Hassel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Jan Leipe
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Matthias Pauschinger
- Department of Cardiology, Klinikum Nürnberg Süd, Paracelsus Medical University Nürnberg, Nuremberg, Germany
| | - Arndt Vogel
- Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital, University Essen-Duisburg, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| |
Collapse
|