1
|
Si F, Ma X, Liu Q, Yu J. Reviewing the path to balance: mechanisms and management of hypertension associated with targeting vascular endothelium in cancer therapy. Hypertens Res 2025:10.1038/s41440-024-02086-8. [PMID: 39820066 DOI: 10.1038/s41440-024-02086-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/13/2024] [Accepted: 12/21/2024] [Indexed: 01/19/2025]
Abstract
Contemporary anticancer drugs are often accompanied by varying degrees of cardiovascular toxicity, with hypertension emerging as one of the most prevalent side effects, particularly linked to inhibitors of vascular endothelial growth factor receptor (VEGFR) and tyrosine kinase inhibitors (TKIs). Hypertension induced by cancer therapies contributes to increased cardiovascular mortality in cancer patients and survivors. Given the shared common risk factors and overlapping pathophysiological mechanisms, hypertension is also a prevalent comorbidity in this patient population. The mechanisms underlying hypertension induced by therapies targeting the vascular endothelial growth factor (VEGF) signaling pathway primarily involve reduced nitric oxide (NO) synthesis, increased endothelin-1 (ET-1) production, oxidative stress, microvascular rarefaction and dysfunction, decreased natriuresis, activation of the renin-angiotensin system (RAS), and partial endothelial cell death. Research into hypertension associated with therapies targeting the VEGF signaling pathway (VSP) could facilitate the optimization of cancer treatments, improve the evaluation and management of hypertension during targeted therapy, and help to reduce cardiovascular event rates and overall patient mortality. This review aims to provide a comprehensive summary of the current advancements in this area.
Collapse
Affiliation(s)
- Fei Si
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xin Ma
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Qian Liu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jing Yu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| |
Collapse
|
2
|
Wegner Wippel C, Deshpande H, Patwa H, Peixoto AJ. Neurogenic orthostatic hypotension after treatment with sorafenib. BMJ Case Rep 2022; 15:e247140. [PMID: 36549761 PMCID: PMC9791444 DOI: 10.1136/bcr-2021-247140] [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] [Accepted: 11/14/2022] [Indexed: 12/24/2022] Open
Abstract
A man in his 70s with a history of fatigue, abdominal pain, and a palpable abdominal mass was found to have a peritoneal desmoid tumour. One year after diagnosis, he was prescribed sorafenib to limit tumour growth. Two months later, he developed dyspnoea on exertion and lower extremity weakness and was reported to have supine hypertension and orthostatic hypotension. On formal autonomic testing, he was noted to have severely impaired sympathetic responses and marked orthostatic hypotension without appropriate chronotropic response. A decision to hold sorafenib was made, and treatment was started with graduated compression stockings, liberal fluid and sodium intake, and midodrine. The patient had a modest and gradual improvement in his symptoms. To our knowledge, this is the first reported case of orthostatic hypotension related to sorafenib or any vascular endothelial growth factor inhibitors.
Collapse
Affiliation(s)
| | - Hari Deshpande
- Medical Oncology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Huned Patwa
- Neurology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Aldo J Peixoto
- Nephrology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
3
|
Chuquin D, Abbate A, Bottinor W. Hypertension in Cancer Survivors: A Review of the Literature and Suggested Approach to Diagnosis and Treatment. J Cardiovasc Pharmacol 2022; 80:522-530. [PMID: 36027586 PMCID: PMC9547865 DOI: 10.1097/fjc.0000000000001342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cardiovascular disease (CVD) is a leading cause of morbidity and mortality among cancer survivors. Hypertension, which is common among cancer survivors with a prevalence of greater than 70% by age 50, potentiates the risk for CVD in a more than additive fashion. For example, childhood cancer survivors who develop hypertension may have up to a 12 times higher risk for heart failure than survivors who remain normotensive. Studies have shown that mild valvular disease (28% incidence), cardiomyopathy (7.4%), arrhythmias (4.6%), and coronary artery disease (3.8%) are among the most common CVDs in childhood cancer survivors. Among adolescent and young adult cancer survivors, the most common reasons for cardiovascular-related hospital admission are venous/lymphatic disease (absolute excess risk 19%), cardiomyopathy and arrhythmia (15%), hypertension (13%), and ischemic heart disease (12%). In addition, cancer therapies can increase the risk for hypertension and CVD. Therefore, early detection and treatment of hypertension is essential to reducing cardiovascular morbidity and mortality among survivors. METHODS We present a literature review, which identified over 20 clinical trials, systemic reviews, and meta-analyses (13 clinical trials, 8 systemic reviews or meta-analyses) by searching PubMed, Google Scholar, and the Cochrane Library for relevant articles addressing hypertension in cancer survivors. RESULTS Although our understanding of the complex relationship between cancer therapies and CVD has grown significantly over the past 2 decades, there remain several gaps in knowledge when specifically addressing CVD in the survivor population. This review provides an up-to-date survivor-centered approach to the screening and treatment of hypertension, which considers survivor-specific cardiovascular risk, applies guideline directed therapies when appropriate, screens for survivor-specific factors that may influence antihypertensive medication selection, and finally considers the prohypertensive mechanisms of antineoplastic agents as a potential target for antihypertensive medications. CONCLUSIONS Screening for and treating hypertension among survivors can promote cardiovascular health in this vulnerable population.
Collapse
Affiliation(s)
- David Chuquin
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| | | | | |
Collapse
|
4
|
Esa R, Steinberg E, Dagan A, Yekhtin Z, Tischenko K, Benny O. Newly synthesized methionine aminopeptidase 2 inhibitor hinders tumor growth. Drug Deliv Transl Res 2022; 13:1170-1182. [PMID: 35637333 DOI: 10.1007/s13346-022-01187-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2022] [Indexed: 11/24/2022]
Abstract
Methionine aminopeptidase 2 (MetAp2) inhibition has been recognized as a promising approach for suppressing angiogenesis and cancer progression. Small molecule fumagillol derivatives with adamantane side groups were synthesized and evaluated for MetAp2 inhibition activity, and a lead molecule with superior abilities to inhibit the enzymatic activity of MetAp2 was identified. The compound, referred to as AD-3281, effectively suppressed proliferation of cancer and endothelial cells and impaired tube formation of endothelial cells in vitro. When administered systemically, AD-3281 was well tolerated and led to a significant suppression of human melanoma and mammary tumor xenografts grown in mice. The activity in vivo was associated with reduced angiogenesis and tumor proliferation as detected histologically. In order to develop a formulation that can solubilize AD-3281 with a minimal content of organic solvents, biodegradable nanoparticles comprised of poly-lactic-co-glycolic acid (PLGA) were fabricated and characterized. Compared with the free compound, AD-3281-loaded nanoparticles showed an advantageous cellular availability and uptake, leading to higher activity in cells and better transport in three-dimensional (3D) cultures. Taken together, we introduce a novel MetAp2 inhibitor with high anti-cancer activity and a stable nano-formulation with a high potential for future clinical translation.
Collapse
Affiliation(s)
- Rawnaq Esa
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Eliana Steinberg
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Arie Dagan
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
- The Lautenberg Center for General and Tumor Immunology, The Hadassah Medical School, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Zhanna Yekhtin
- The Lautenberg Center for General and Tumor Immunology, The Hadassah Medical School, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Katerina Tischenko
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Ofra Benny
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel.
| |
Collapse
|
5
|
Hypoxia signaling and oxygen metabolism in cardio-oncology. J Mol Cell Cardiol 2022; 165:64-75. [PMID: 34979102 DOI: 10.1016/j.yjmcc.2021.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/05/2021] [Accepted: 12/27/2021] [Indexed: 12/26/2022]
Abstract
Cardio-oncology is a rapidly growing field in cardiology that focuses on the management of cardiovascular toxicities associated with cancer-directed therapies. Tumor hypoxia is a central driver of pathologic tumor growth, metastasis, and chemo-resistance. In addition, conditions that mimic hypoxia (pseudo-hypoxia) play a causal role in the pathogenesis of numerous types of cancer, including renal cell carcinoma. Therefore, therapies targeted at hypoxia signaling pathways have emerged over the past several years. Though efficacious, these therapies are associated with significant cardiovascular toxicities, ranging from hypertension to cardiomyopathy. This review focuses on oxygen metabolism in tumorigenesis, the role of targeting hypoxia signaling in cancer therapy, and the relevance of oxygen metabolism in cardio-oncology. This review will specifically focus on hypoxia signaling mediated by hypoxia-inducible factors and the prolyl hydroxylase oxygen-sensing enzymes, the cardiovascular effects of specific cancer targeted therapies mediated on VEGF and HIF signaling, hypoxic signaling in cardiovascular disease, and the role of oxygen in anthracycline cardiotoxicity. The implications of these therapies on myocardial biology and cardiac function are discussed, underlining the fine balance of hypoxia signaling in cardiac homeostasis. Understanding these cardiovascular toxicities will be important to optimize treatment for cancer patients while mitigating potentially severe cardiovascular side effects.
Collapse
|
6
|
Mandry D, Girerd N, Lamiral Z, Huttin O, Filippetti L, Micard E, Ncho Mottoh MPB, Böhme P, Chemla D, Zannad F, Rossignol P, Marie PY. Arterial and Cardiac Remodeling Associated With Extra Weight Gain in an Isolated Abdominal Obesity Cohort. Front Cardiovasc Med 2021; 8:771022. [PMID: 34805324 PMCID: PMC8602697 DOI: 10.3389/fcvm.2021.771022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022] Open
Abstract
Introduction: This study aims to assess the changes in cardiovascular remodeling attributable to bodyweight gain in a middle-aged abdominal obesity cohort. A remodeling worsening might explain the increase in cardiovascular risk associated with a dynamic of weight gain. Methods: Seventy-five middle-aged subjects (56 ± 5 years, 38 women) with abdominal obesity and no known cardiovascular disease underwent MRI-based examinations at baseline and at a 6.1 ± 1.2-year follow-up to monitor cardiovascular remodeling and hemodynamic variables, most notably the effective arterial elastance (Ea). Ea is a proxy of the arterial load that must be overcome during left ventricular (LV) ejection, with increased EA resulting in concentric LV remodeling. Results: Sixteen obese subjects had significant weight gain (>7%) during follow-up (WG+), whereas the 59 other individuals did not (WG-). WG+ and WG- exhibited significant differences in the baseline to follow-up evolutions of several hemodynamic parameters, notably diastolic and mean blood pressures (for mean blood pressure, WG+: +9.3 ± 10.9 mmHg vs. WG-: +1.7 ± 11.8 mmHg, p = 0.022), heart rate (WG+: +0.6 ± 9.4 min-1 vs. -8.9 ± 11.5 min-1, p = 0.003), LV concentric remodeling index (WG: +0.08 ± 0.16 g.mL-1 vs. WG-: -0.02 ± 0.13 g.mL-1, p = 0.018) and Ea (WG+: +0.20 ± 0.28 mL mmHg-1 vs. WG-: +0.01 ± 0.30 mL mmHg-1, p = 0.021). The evolution of the LV concentric remodeling index and Ea were also strongly correlated in the overall obese population (p < 0.001, R2 = 0.31). Conclusions: A weight gain dynamic is accompanied by increases in arterial load and load-related concentric LV remodeling in an isolated abdominal obesity cohort. This remodeling could have a significant impact on cardiovascular risk.
Collapse
Affiliation(s)
- Damien Mandry
- Department of Radiology, CHRU-Nancy, Université de Lorraine, Nancy, France.,INSERM, UMR-1254, Université de Lorraine, Nancy, France
| | - Nicolas Girerd
- INSERM, UMR-1116, Université de Lorraine, Nancy, France.,Department of Cardiology, CHRU-Nancy, Université de Lorraine, Nancy, France.,CHRU-Nancy, INSERM, CIC 1433, Université de Lorraine, Nancy, France
| | - Zohra Lamiral
- CHRU-Nancy, INSERM, CIC 1433, Université de Lorraine, Nancy, France
| | - Olivier Huttin
- INSERM, UMR-1116, Université de Lorraine, Nancy, France.,Department of Cardiology, CHRU-Nancy, Université de Lorraine, Nancy, France
| | - Laura Filippetti
- Department of Cardiology, CHRU-Nancy, Université de Lorraine, Nancy, France
| | - Emilien Micard
- CHRU-Nancy, INSERM, CIC 1433, Université de Lorraine, Nancy, France
| | | | - Philip Böhme
- Department of Endocrinology, Diabetology, Nutrition, CHRU-Nancy, Nancy, France
| | - Denis Chemla
- Explorations Fonctionnelles, Hôpital Kremlin Bicêtre, APHP, Paris, France.,INSERM, UMR- 999, Hôpital Marie-Lannelongue, Le Plessis-Robinson, France
| | - Faïez Zannad
- INSERM, UMR-1116, Université de Lorraine, Nancy, France.,Department of Cardiology, CHRU-Nancy, Université de Lorraine, Nancy, France.,CHRU-Nancy, INSERM, CIC 1433, Université de Lorraine, Nancy, France
| | - Patrick Rossignol
- INSERM, UMR-1116, Université de Lorraine, Nancy, France.,CHRU-Nancy, INSERM, CIC 1433, Université de Lorraine, Nancy, France.,FCRIN INI-CRCT, Nancy, France
| | - Pierre-Yves Marie
- INSERM, UMR-1116, Université de Lorraine, Nancy, France.,CHRU-Nancy, Université de Lorraine, Nuclear Medicine & Nancyclotep Platform, Nancy, France
| |
Collapse
|
7
|
The vascular endothelial growth factor trap aflibercept induces vascular dysfunction and hypertension via attenuation of eNOS/NO signaling in mice. Acta Pharmacol Sin 2021; 42:1437-1448. [PMID: 33303990 PMCID: PMC8379246 DOI: 10.1038/s41401-020-00569-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022] Open
Abstract
Aflibercept, as a soluble decoy vascular endothelial growth factor receptor, Which has been used as a first-line monotherapy for cancers. Aflibercept often causes cardiovascular toxicities including hypertension, but the mechanisms underlying aflibercept-induced hypertension remain unknown. In this study we investigated the effect of short-term and long-term administration of aflibercept on blood pressure (BP), vascular function, NO bioavailability, oxidative stress and endothelin 1 (ET-1) in mice and cultured endothelial cells. We showed that injection of a single-dose of aflibercept (18.2, 36.4 mg/kg, iv) rapidly and dose-dependently elevated BP in mice. Aflibercept treatment markedly impaired endothelial-dependent relaxation (EDR) and resulted in NADPH oxidases 1 (NOX1)- and NADPH oxidases 4 (NOX4)-mediated generation of ROS, decreased the activation of protein kinase B (Akt) and endothelial nitric oxide synthase (eNOS) concurrently with a reduction in nitric oxide (NO) production and elevation of ET-1 levels in mouse aortas; these effects were greatly attenuated by supplementation of L-arginine (L-arg, 0.5 or 1.0 g/kg, bid, ig) before aflibercept injection. Similar results were observed in L-arg-pretreated cultured endothelial cells, showing markedly decreased ROS accumulation and AKT/eNOS/NO signaling impairment induced by aflibercept. In order to assess the effects of long-term aflibercept on hypertension and to evaluate the beneficial effects of L-arg supplementation, we administered these two drugs to WT mice for up to 14 days (at an interval of two days). Long-term administration of aflibercept resulted in a sustained increase in BP and a severely impaired EDR, which are associated with NOX1/NOX4-mediated production of ROS, increase in ET-1, inhibition of AKT/eNOS/NO signaling and a decreased expression of cationic amino acid transporter (CAT-1). The effects caused by long-term administration were greatly attenuated by L-arg supplementation in a dose-dependent manner. We conclude that aflibercept leads to vascular dysfunction and hypertension by inhibiting CAT-1/AKT/eNOS/NO signaling, increasing ET-1, and activating NOX1/NOX4-mediated oxidative stress, which can be suppressed by supplementation of L-arg. Therefore, L-arg could be a potential therapeutic agent for aflibercept-induced hypertension.
Collapse
|
8
|
Pepe GJ, Albrecht ED. Novel Technologies for Target Delivery of Therapeutics to the Placenta during Pregnancy: A Review. Genes (Basel) 2021; 12:1255. [PMID: 34440429 PMCID: PMC8392549 DOI: 10.3390/genes12081255] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023] Open
Abstract
Uterine spiral artery remodeling is essential for placental perfusion and fetal growth and, when impaired, results in placental ischemia and pregnancy complications, e.g., fetal growth restriction, preeclampsia, premature birth. Despite the high incidence of adverse pregnancies, current treatment options are limited. Accordingly, research has shifted to the development of gene therapy technologies that provide targeted delivery of "payloads" to the placenta while limiting maternal and fetal exposure. This review describes the current strategies, including placental targeting peptide-bound liposomes, nanoparticle or adenovirus constructs decorated with specific peptide sequences and placental gene promoters delivered via maternal IV injection, directly into the placenta or the uterine artery, as well as noninvasive site-selective targeting of regulating genes conjugated with microbubbles via contrast-enhanced ultrasound. The review also provides a perspective on the effectiveness of these technologies in various animal models and their practicability and potential use for targeted placental delivery of therapeutics and genes in adverse human pregnancies affected by placental dysfunction.
Collapse
Affiliation(s)
- Gerald J. Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA;
| | - Eugene D. Albrecht
- Departments of Obstetrics/Gynecology/Reproductive Sciences and Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
9
|
Hahn VS, Zhang KW, Sun L, Narayan V, Lenihan DJ, Ky B. Heart Failure With Targeted Cancer Therapies: Mechanisms and Cardioprotection. Circ Res 2021; 128:1576-1593. [PMID: 33983833 DOI: 10.1161/circresaha.121.318223] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oncology has seen growing use of newly developed targeted therapies. Although this has resulted in dramatic improvements in progression-free and overall survival, challenges in the management of toxicities related to longer-term treatment of these therapies have also become evident. Although a targeted approach often exploits the differences between cancer cells and noncancer cells, overlap in signaling pathways necessary for the maintenance of function and survival in multiple cell types has resulted in systemic toxicities. In particular, cardiovascular toxicities are of important concern. In this review, we highlight several targeted therapies commonly used across a variety of cancer types, including HER2 (human epidermal growth factor receptor 2)+ targeted therapies, tyrosine kinase inhibitors, immune checkpoint inhibitors, proteasome inhibitors, androgen deprivation therapies, and MEK (mitogen-activated protein kinase kinase)/BRAF (v-raf murine sarcoma viral oncogene homolog B) inhibitors. We present the oncological indications, heart failure incidence, hypothesized mechanisms of cardiotoxicity, and potential mechanistic rationale for specific cardioprotective strategies.
Collapse
Affiliation(s)
- Virginia S Hahn
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD (V.S.H.)
| | - Kathleen W Zhang
- Cardio-Oncology Center of Excellence, Washington University, St Louis, MO (K.W.Z., D.J.L.)
| | - Lova Sun
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Vivek Narayan
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniel J Lenihan
- Cardio-Oncology Center of Excellence, Washington University, St Louis, MO (K.W.Z., D.J.L.)
| | - Bonnie Ky
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Division of Cardiovascular Medicine (B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Division of Biostatistics (B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
10
|
Neves KB, Montezano AC, Lang NN, Touyz RM. Vascular toxicity associated with anti-angiogenic drugs. Clin Sci (Lond) 2020; 134:2503-2520. [PMID: 32990313 DOI: 10.1042/cs20200308] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Over the past two decades, the treatment of cancer has been revolutionised by the highly successful introduction of novel molecular targeted therapies and immunotherapies, including small-molecule kinase inhibitors and monoclonal antibodies that target angiogenesis by inhibiting vascular endothelial growth factor (VEGF) signaling pathways. Despite their anti-angiogenic and anti-cancer benefits, the use of VEGF inhibitors (VEGFi) and other tyrosine kinase inhibitors (TKIs) has been hampered by potent vascular toxicities especially hypertension and thromboembolism. Molecular processes underlying VEGFi-induced vascular toxicities still remain unclear but inhibition of endothelial NO synthase (eNOS), reduced nitric oxide (NO) production, oxidative stress, activation of the endothelin system, and rarefaction have been implicated. However, the pathophysiological mechanisms still remain elusive and there is an urgent need to better understand exactly how anti-angiogenic drugs cause hypertension and other cardiovascular diseases (CVDs). This is especially important because VEGFi are increasingly being used in combination with other anti-cancer dugs, such as immunotherapies (immune checkpoint inhibitors (ICIs)), other TKIs, drugs that inhibit epigenetic processes (histone deacetylase (HDAC) inhibitor) and poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors, which may themselves induce cardiovascular injury. Here, we discuss vascular toxicities associated with TKIs, especially VEGFi, and provide an up-to-date overview on molecular mechanisms underlying VEGFi-induced vascular toxicity and cardiovascular sequelae. We also review the vascular effects of VEGFi when used in combination with other modern anti-cancer drugs.
Collapse
Affiliation(s)
- Karla B Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Ninian N Lang
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| |
Collapse
|
11
|
Bottinor W, Parikh A, Jahangir E. Emerging cancer therapies and cardiovascular risk. J Thromb Thrombolysis 2020; 51:837-845. [PMID: 32886244 DOI: 10.1007/s11239-020-02263-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The cardiovascular (CV) toxicity profiles of traditional cancer therapies such as anthracyclines and radiation therapy are familiar to many cardiologists. With the development and widespread use of additional cancer therapeutics, CV toxicities related to these agents are becoming more common. Cardiovascular specialists are often integrated into the care team for individuals with cancer and knowledge of the CV toxicities of cancer therapeutics has become essential. In this review, we provide a clinically focused summary of the current data regarding CV toxicities of common cancer therapies and identify potential management strategies for the CV specialist.
Collapse
Affiliation(s)
- Wendy Bottinor
- Division of Cardiovascular Medicine, Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Gateway bldg. 1200 E Marshall St, Richmond, VA, 23298, USA. .,Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Amar Parikh
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eiman Jahangir
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
12
|
Mechanism of Vascular Toxicity in Rats Subjected to Treatment with a Tyrosine Kinase Inhibitor. TOXICS 2020; 8:toxics8030049. [PMID: 32698382 PMCID: PMC7560282 DOI: 10.3390/toxics8030049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/17/2020] [Indexed: 12/01/2022]
Abstract
Sunitinib (Su) is a tyrosine kinase inhibitor with antiangiogenic and antineoplastic effects that is recommended therapy for renal cell carcinoma, gastrointestinal stromal tumors, and pancreatic neuroendocrine tumors. Arterial hypertension is one of the adverse effects observed in the treatment with Su. The aim of this work was to deepen our understanding of the underlying mechanisms involved in the development of this side effect. Studies on endothelial function, vascular remodeling and nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase) system were carried out in thoracic aortas from rats treated with Su for three weeks. Animals subjected to Su treatment presented with increased blood pressure and reduced endothelium-dependent vasodilation, the latter being reverted by NADPH oxidase blockade. Furthermore, vascular remodeling and stronger Masson trichrome staining, together with enhanced immunofluorescence signal for collagen 1 alpha 1 (Col1α1), were observed in aortas from treated animals. These results were accompanied by a significant elevation in superoxide anion production and the activity/protein/gene expression of NADPH oxidase isoforms (NOX1, NOX2, and NOX4), which was also prevented by NOX inhibition. Furthermore, a decrease in nitric oxide (NO) levels and endothelial nitric oxide synthase (eNOS) activation was observed in aortas from Su-treated animals. All these results indicate that endothelial dysfunction secondary to changes in vascular remodeling and oxidative stress might be responsible for the typical arterial hypertension that develops following treatment with Su.
Collapse
|
13
|
Abstract
BACKGROUND Cardio-oncology aims to mitigate adverse cardiovascular manifestations in cancer survivors, but treatment-induced hypertension or aggravated hypertension has received less attention in these high cardiovascular risk patients. METHODS In this systematic review, we searched literature for contemporary data on the prevalence, pathophysiologic mechanisms, treatment implications and preventive strategies of hypertension in patients under antineoplastic therapy. RESULTS Several classes of antineoplastic drugs, including mainly vascular endothelial growth factor inhibitors, proteasome inhibitors, cisplatin derivatives, corticosteroids or radiation therapy were consistently associated with increased odds for new-onset hypertension or labile hypertensive status in previous controlled patients. Moreover, hypertension constitutes a major risk factor for chemotherapy-induced cardiotoxicity, which is the most serious cardiovascular adverse effect of antineoplastic therapy. Despite the heterogeneity of pooled studies, the pro-hypertensive profile of examined drug classes could be attributed to common structural and functional disorders. Importantly, certain antihypertensive drugs are considered to be more effective in the management of hypertension in this population and may partially attenuate indirect complications of cancer treatment, such as progressive development of cardiomyopathy and/or cardiovascular death. Nonpharmacological approaches to alleviate hypertension in cancer patients are also described, albeit adjudicated as less effective in general. CONCLUSION A growing body of evidence suggests that multiple antineoplastic agents increase the rate of progression of hypertension. Physicians need to balance the life-saving cancer treatment and the inflated risk of adverse cardiovascular events due to suboptimal management of hypertension in order to achieve improved clinical outcomes and sustained survival for their patients.
Collapse
|
14
|
Sonaglioni A, Albini A, Fossile E, Pessi MA, Nicolosi GL, Lombardo M, Anzà C, Ambrosio G. Speckle-Tracking Echocardiography for Cardioncological Evaluation in Bevacizumab-Treated Colorectal Cancer Patients. Cardiovasc Toxicol 2020; 20:581-592. [PMID: 32519318 DOI: 10.1007/s12012-020-09583-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Angiogenesis inhibitor Bevacizumab (BVZ) may lead to the development of adverse effects, including hypertension and cardiac ischemia. Whether assessment of changes in myocardial strain by two-dimensional speckle-tracking echocardiography (2D-STE) can be of value in detecting BVZ-mediated cardiotoxicity at an earlier stage is not known. We investigated whether 2D-STE can non-invasively detect early evidence of cardiotoxicity in metastatic colorectal cancer (mCRC) patients treated with BVZ. Between January and June 2019, 25 consecutive patients (71.8 ± 7.5 year/old, 17 males) with mCRC were prospectively enrolled. Patients underwent physical examination, blood tests, and conventional 2D-transthoracic echocardiography implemented with 2D-STE analysis, at baseline and at 3 and 6 months following treatment with BVZ (15 mg/kg every 15 days) + 5-fluorouracil/folinic acid plus oxaliplatin (FOLFOX i.v.). At 6-month follow-up, we assessed occurrence of global longitudinal strain (GLS) impairment (> 15% decrease in GLS compared with baseline) as primary end-point and a new-onset systemic hypertension (secondary end-point). On average, GLS showed a progressive significant impairment after BVZ, from - 17.4 ± 3.2% at baseline to - 16 ± 2.9% (p = 0.003) at 6-month follow-up; > 15% decrease in GLS (primary end-point) was detected in 9 patients (36%). All other strain parameters remained unchanged. New-onset systemic hypertension (secondary end-point) was diagnosed in five patients (20%). No significant changes were observed in serial high-sensitivity cardiac troponin I measurements. No patient developed significant changes in LV size or LV ejection fraction; no case of clinically symptomatic HF was observed during BVZ-treatment. Measurement of GLS by 2D-STE analysis can effectively detect BVZ-mediated cardiotoxicity at an early stage.
Collapse
Affiliation(s)
- Andrea Sonaglioni
- Department of Cardiology, Ospedale San Giuseppe IRCCS MultiMedica, Milan, Italy
| | - Adriana Albini
- Scientific and Technological Pole, IRCCS MultiMedica, Milan, Italy.
| | - Emanuela Fossile
- Department of Oncology, Ospedale San Giuseppe IRCCS MultiMedica, Milan, Italy
| | | | | | - Michele Lombardo
- Department of Cardiology, Ospedale San Giuseppe IRCCS MultiMedica, Milan, Italy
| | - Claudio Anzà
- Cardiovascular Department, IRCCS MultiMedica, Sesto San Giovanni (MI), Italy
| | - Giuseppe Ambrosio
- Cardiology and Cardiovascular Pathophysiology, Azienda Ospedaliero-Universitaria "S. Maria Della Misericordia", Perugia, Italy
| |
Collapse
|
15
|
Bottinor WJ, Shuey MM, Manouchehri A, Farber-Eger EH, Xu M, Nair D, Salem JE, Wang TJ, Brittain EL. Renin-Angiotensin-Aldosterone System Modulates Blood Pressure Response During Vascular Endothelial Growth Factor Receptor Inhibition. JACC: CARDIOONCOLOGY 2019; 1:14-23. [PMID: 32984850 PMCID: PMC7513950 DOI: 10.1016/j.jaccao.2019.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Objectives This study postulated that antihypertensive therapy with renin-angiotensin-aldosterone system (RAAS) inhibition may mitigate vascular endothelial growth factor inhibitor (VEGFi)–mediated increases in blood pressure more effectively than other antihypertensive medications in patients receiving VEGFi therapy. Background VEGFi therapy is commonly used in the treatment of cancer. One common side effect of VEGFi therapy is elevated blood pressure. Evidence suggests that the RAAS may be involved in VEGFi-mediated increases in blood pressure. Methods This retrospective cohort analysis was performed using a de-identified version of the electronic health record at Vanderbilt University Medical Center in Nashville, Tennessee. Subjects with cancer who were exposed to VEGFi therapy were identified, and blood pressure and medication data were extracted. Changes in mean systolic and diastolic blood pressure in response to VEGFi therapy in patients receiving RAAS inhibitor (RAASi) therapy before VEGFi initiation were compared with changes in mean systolic and diastolic blood pressure in patients not receiving RAASi therapy before VEGFi initiation. Results Mean systolic and diastolic blood pressure rose in both groups after VEGFi use; however, patients who had RAASi therapy before VEGFi initiation had a significantly lower increase in systolic blood pressure as compared with patients with no RAASi therapy (2.46 mm Hg [95% confidence interval: 0.7 to 4.2] compared with 4.56 mm Hg [95% confidence interval: 3.5 to 5.6], respectively; p = 0.034). Conclusions In a real-world clinical population, RAASi therapy before VEGFi initiation may ameliorate VEGFi-mediated increases in blood pressure. Randomized clinical trials are needed to further our understanding of the role of RAASi therapy in VEGFi-mediated increases in blood pressure.
Collapse
Affiliation(s)
- Wendy J Bottinor
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Megan M Shuey
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ali Manouchehri
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Eric H Farber-Eger
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Meng Xu
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee
| | - Devika Nair
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joe-Elie Salem
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee.,Department of Clinical Pharmacology, University of the Sorbonne, Assistance Publique Hôpitaux de Paris, Institut National de la Santé et de la Recherche Médicale CIC 14-21, Pitié-Salpêtrière Hospital, Paris, France
| | - Thomas J Wang
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Evan L Brittain
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
16
|
Affiliation(s)
- Rhian M. Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Ninian N. Lang
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| |
Collapse
|
17
|
Babischkin JS, Aberdeen GW, Lindner JR, Bonagura TW, Pepe GJ, Albrecht ED. Vascular Endothelial Growth Factor Delivery to Placental Basal Plate Promotes Uterine Artery Remodeling in the Primate. Endocrinology 2019; 160:1492-1505. [PMID: 31002314 PMCID: PMC6542484 DOI: 10.1210/en.2019-00059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
Extravillous trophoblast (EVT) uterine artery remodeling (UAR) promotes placental blood flow, but UAR regulation is unproven. Elevating estradiol (E2) in early baboon pregnancy suppressed UAR and EVT vascular endothelial growth factor (VEGF) expression, but this did not prove that VEGF mediated this process. Therefore, our primate model of prematurely elevating E2 and contrast-enhanced ultrasound cavitation of microbubble (MB) carriers was used to deliver VEGF DNA to the placental basal plate (PBP) to establish the role of VEGF in UAR. Baboons were treated on days 25 to 59 of gestation (term, 184 days) with E2 alone or with E2 plus VEGF DNA-conjugated MBs briefly infused via a maternal peripheral vein on days 25, 35, 45, and 55. At each of these times an ultrasound beam was directed to the PBP to collapse the MBs and release VEGF DNA. VEGF DNA-labeled MBs per contrast agent was localized in the PBP but not the fetus. Remodeling of uterine arteries >25 µm in diameter on day 60 was 75% lower (P < 0.001) in E2-treated (7% ± 2%) than in untreated baboons (30% ± 4%) and was restored to normal by E2/VEGF. VEGF protein levels (signals/nuclear area) within the PBP were twofold lower (P < 0.01) in E2-treated (4.2 ± 0.9) than in untreated (9.8 ± 2.8) baboons and restored to normal by E2/VEGF (11.9 ± 1.6), substantiating VEGF transfection. Thus, VEGF gene delivery selectively to the PBP prevented the decrease in UAR elicited by prematurely elevating E2 levels, establishing the role of VEGF in regulating UAR in vivo during primate pregnancy.
Collapse
Affiliation(s)
- Jeffery S Babischkin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Graham W Aberdeen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, Oregon
| | | | - Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Eugene D Albrecht
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
- Correspondence: Eugene D. Albrecht, PhD, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Bressler Research Laboratories 11-019, 655 West Baltimore Street, Baltimore, Maryland 21201. E-mail:
| |
Collapse
|
18
|
Nguyen T, Davidson BP. Contrast Enhanced Ultrasound Perfusion Imaging in Skeletal Muscle. J Cardiovasc Imaging 2019; 27:163-177. [PMID: 31161755 PMCID: PMC6669180 DOI: 10.4250/jcvi.2019.27.e31] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/21/2019] [Indexed: 12/14/2022] Open
Abstract
The ability to accurately evaluate skeletal muscle microvascular blood flow has broad clinical applications for understanding the regulation of skeletal muscle perfusion in health and disease states. Contrast-enhanced ultrasound (CEU) perfusion imaging, a technique originally developed to evaluate myocardial perfusion, is one of many techniques that have been applied to evaluate skeletal muscle perfusion. Among the advantages of CEU perfusion imaging of skeletal muscle is that it is rapid, safe and performed with equipment already present in most vascular medicine laboratories. The aim of this review is to discuss the use of CEU perfusion imaging in skeletal muscle. This article provides details of the protocols for CEU imaging in skeletal muscle, including two predominant methods for bolus and continuous infusion destruction-replenishment techniques. The importance of stress perfusion imaging will be highlighted, including a discussion of the methods used to produce hyperemic skeletal muscle blood flow. A broad overview of the disease states that have been studied in humans using CEU perfusion imaging of skeletal muscle will be presented including: (1) peripheral arterial disease; (2) sickle cell disease; (3) diabetes; and (4) heart failure. Finally, future applications of CEU imaging in skeletal muscle including therapeutic CEU imaging will be discussed along with technological developments needed to advance the field.
Collapse
Affiliation(s)
- TheAnh Nguyen
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Brian P Davidson
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA.,Veterans Affairs Portland Health Care System, Portland, OR, USA.
| |
Collapse
|
19
|
Yu BB, Zhi H, Zhang XY, Liang JW, He J, Su C, Xia WH, Zhang GX, Tao J. Mitochondrial dysfunction-mediated decline in angiogenic capacity of endothelial progenitor cells is associated with capillary rarefaction in patients with hypertension via downregulation of CXCR4/JAK2/SIRT5 signaling. EBioMedicine 2019; 42:64-75. [PMID: 30904607 PMCID: PMC6491423 DOI: 10.1016/j.ebiom.2019.03.031] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 01/04/2023] Open
Abstract
Background Hypertensive patients exhibit decline in capillary density and endothelial progenitor cells (EPCs). However, whether capillary rarefaction in hypertension is associated with defect angiogenesis of EPCs remains unknown. We hypothesized that impaired mitochondrial function of late EPCs in hypertension is associated with the structural lack of capillary microcirculation via deficient CXCR4/JAK2/SIRT5 signaling. Methods We performed capillary microcirculation detection in hypertensive patients and healthy subjects. Angiogenic capacity and mitochondrial function of circulating EPCs were evaluated. The underlying mechanisms were further investigated by genetic inhibition and overexpression. Findings Capillary density of nail fold and eye fundus were significantly reduced in hypertensive patients, which was paralleled to decreased in vitro late EPC function and in vivo angiogenic capacity. Meanwhile the decline of EPC function in hypertension was accompanied by impaired mitochondrial ultrastructure, diminished mitochondrial membrane potential, reduced oxygen consumption, increased ROS generation and NADH level. Rotenone induced inhibition of oxygen consumption rate, excessive ROS generation and loss of MMP, which markedly decreased the in vitro functions of EPCs. Furthermore, SIRT5 expression of EPCs in hypertension was markedly reduced, which was correlated to mitochondrial dysfunction. CXCR4 gene transfer enhanced SIRT5 expression, improved mitochondrial functions and augmented angiogenic capacity of EPCs. The beneficial impacts of SIRT5 up-regulation on late EPC-mediated angiogenesis can be abrogated by blockade of CXCR4/JAK2/SIRT5 signaling pathway. Interpretation Mitochondrial dysfunction-mediated fall in angiogenic capacity due to deficient CXCR4/JAK2/SIRT5 signaling of late EPCs is probably responsible for the capillary rarefaction in hypertension. Our findings provide insight into the potential of EPC mitochondria as a novel target for the treatment of hypertension-related loss of microvascular density. Funds National Nature Science Foundation of China, 973Program, the Nature Science Foundation of Guangdong.
Collapse
Affiliation(s)
- Bing-Bo Yu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, China
| | - Hui Zhi
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, China
| | - Xiao-Yu Zhang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, China
| | - Jian-Wen Liang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, China
| | - Jiang He
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, China
| | - Chen Su
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, China
| | - Wen-Hao Xia
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, China
| | - Gao-Xing Zhang
- Department of Cardiovascular Disease, The Jiangmen Central Hospital, China.
| | - Jun Tao
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, China.
| |
Collapse
|
20
|
Mozolevska V, Schwartz A, Cheung D, Goyal V, Shaikh B, Dingman B, Kim E, Mittal I, Asselin CY, Edel A, Ravandi A, Thliveris J, Singal PK, Czaykowski P, Jassal DS. Role of renin-angiotensin system antagonists in the prevention of bevacizumab- and sunitinib-mediated cardiac dysfunction. Am J Physiol Heart Circ Physiol 2019; 316:H446-H458. [DOI: 10.1152/ajpheart.00344.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although anticancer systemic therapy agents clearly lead to improved survival in patients with cancer, these can come at the cost of serious complications including cardiotoxicity. Two types of targeted systemic therapies currently in use for colorectal cancer (CRC) and renal cell cancer (RCC), respectively, include the vascular endothelial growth factor inhibitor bevacizumab (BVZ) and the tyrosine kinase inhibitor sunitinib (SNT). Despite the beneficial effects of BVZ and SNT in improving clinical outcomes in the settings of CRC and RCC, there is an increased risk of cardiac dysfunction. The aim of the present study was to determine whether prophylactic administration of renin-angiotensin system (RAS) inhibitors would attenuate the cardiotoxic side effects of BVZ or SNT in a chronic in vivo murine model. A total of 194 wild-type C57Bl/6 male mice received: 1) 0.9% saline, 2) BVZ (10 mg·kg−1·wk−1), or 3) SNT (40 mg·kg−1·day−1) for 4 wk. Within each arm, mice received daily prophylactic treatment with hydralazine (0.05 mg/ml), aliskiren (50 mg/kg), perindopril (4 mg/kg), or valsartan (2 mg/kg). Although hydralazine effectively lowered blood pressure in BVZ- or SNT-treated mice, it did not prevent left ventricular systolic dysfunction. Prophylactic administration of aliskiren, perindopril, or valsartan prevented adverse cardiovascular remodeling in mice treated with either BVZ or SNT. The addition of RAS antagonists also downregulated expression of phosphorylated p38 and Bcl-2-like 19-kDa interacting protein 3 in SNT-treated mice. In our chronic in vivo murine model, RAS antagonists partially attenuated the development of BVZ- or SNT-mediated cardiac dysfunction. Future clinical studies are warranted to investigate the cardioprotective effects of prophylactic treatment with RAS inhibitors in the settings of CRC and RCC. NEW & NOTEWORTHY In the evolving field of cardio-oncology, bevacizumab and sunitinib improve clinical outcomes in the settings of metastatic colorectal cancer and renal cell cancer, respectively. These anticancer drugs, however, are associated with an increased risk of cardiotoxicity. The prophylactic administration of renin-angiotensin system antagonists is partially cardioprotective against bevacizumab- and sunitinib-mediated cardiac dysfunction.
Collapse
Affiliation(s)
- Viktoriya Mozolevska
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anna Schwartz
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Cheung
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Vineet Goyal
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Bilal Shaikh
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Bella Dingman
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Esther Kim
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ishika Mittal
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Chantal Y. Asselin
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrea Edel
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Amir Ravandi
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James Thliveris
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pawan K. Singal
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Piotr Czaykowski
- Section of Hematology/Oncology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Davinder S. Jassal
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Section of Hematology/Oncology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Radiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
21
|
Nhola LF, Abdelmoneim SS, Villarraga HR, Kohli M, Grothey A, Bordun KA, Cheung M, Best R, Cheung D, Huang R, Barros-Gomes S, Pitz M, Singal PK, Jassal DS, Mulvagh SL. Echocardiographic Assessment for the Detection of Cardiotoxicity Due to Vascular Endothelial Growth Factor Inhibitor Therapy in Metastatic Renal Cell and Colorectal Cancers. J Am Soc Echocardiogr 2018; 32:267-276. [PMID: 30459123 DOI: 10.1016/j.echo.2018.09.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cardio-oncology is a recently established discipline that focuses on the management of patients with cancer who are at risk for developing cardiovascular complications as a result of their underlying oncologic treatment. In metastatic colorectal cancer (mCRC) and metastatic renal cell carcinoma (mRCC), vascular endothelial growth factor inhibitor (VEGF-i) therapy is commonly used to improve overall survival. Although these novel anticancer drugs may lead to the development of cardiotoxicity, whether early detection of cardiac dysfunction using serial echocardiography could potentially prevent the development of heart failure in this patient population requires further study. The aim of this study was to investigate the role of two-dimensional speckle-tracking echocardiography in the detection of cardiotoxicity due to VEGF-i therapy in patients with mCRC or mRCC. METHODS Patients with mRCC or mCRC were evaluated using serial echocardiography at baseline and 1, 3, and 6 months following VEGF-i treatment. RESULTS A total of 40 patients (34 men; mean age, 63 ± 9 years) receiving VEGF-i therapy were prospectively recruited at two academic centers: 26 (65%) were receiving sunitinib, eight (20%) pazopanib, and six (15%) bevacizumab. The following observations were made: (1) 8% of patients developed clinically asymptomatic cancer therapeutics-related cardiac dysfunction; (2) 30% of patients developed clinically significant decreases in global longitudinal strain, a marker for early subclinical cardiac dysfunction; (3) baseline abnormalities in global longitudinal strain may identify a subset of patients at higher risk for developing cancer therapeutics-related cardiac dysfunction; and (4) new or worsening hypertension was the most common adverse cardiovascular event, afflicting nearly one third of the study population. CONCLUSIONS Cardiac dysfunction defined by serial changes in myocardial strain assessed using two-dimensional speckle-tracking echocardiography occurs in patients undergoing treatment with VEGF-i for mCRC or mRCC, which may provide an opportunity for preventive interventions.
Collapse
Affiliation(s)
- Lara F Nhola
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sahar S Abdelmoneim
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota; Division of Cardiology, Orman Heart Center, Assiut University, Assiut, Egypt
| | | | - Manish Kohli
- Department of Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Axel Grothey
- Department of Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kimberly-Ann Bordun
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Matthew Cheung
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ryan Best
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Cheung
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Runqing Huang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Marshall Pitz
- Section of Hematology/Oncology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pawan K Singal
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Davinder S Jassal
- Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Section of Hematology/Oncology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Section of Cardiology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sharon L Mulvagh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota; Division of Cardiology, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
22
|
Touyz RM, Herrmann J. Cardiotoxicity with vascular endothelial growth factor inhibitor therapy. NPJ Precis Oncol 2018; 2:13. [PMID: 30202791 PMCID: PMC5988734 DOI: 10.1038/s41698-018-0056-z] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis inhibitors targeting the vascular endothelial growth factor (VEGF) signaling pathway (VSP) have been important additions in the therapy of various cancers, especially renal cell carcinoma and colorectal cancer. Bevazicumab, the first VSP to receive FDA approval in 2004 targeting all circulating isoforms of VEGF-A, has become one of the best-selling drugs of all times. The second wave of tyrosine kinase inhibitors (TKIs), which target the intracellular site of VEGF receptor kinases, began with the approval of sorafenib in 2005 and sunitinib in 2006. Heart failure was subsequently noted, in 2-4% of patients on bevacizumab and in 3-8% of patients on VSP-TKIs. The very fact that the single-targeted monoclonal antibody bevacizumab can induce cardiotoxicity supports a pathomechanistic role for the VSP and the postulate of the "vascular" nature of VSP inhibitor cardiotoxicity. In this review we will outline this scenario in greater detail, reflecting on hypertension and coronary artery disease as risk factors for VSP inhibitor cardiotoxicity, but also similarities with peripartum and diabetic cardiomyopathy. This leads to the concept that any preexisting or coexisting condition that reduces the vascular reserve or utilizes the vascular reserve for compensatory purposes may pose a risk factor for cardiotoxicity with VSP inhibitors. These conditions need to be carefully considered in cancer patients who are to undergo VSP inhibitor therapy. Such vigilance is not to exclude patients from such prognostically extremely important therapy but to understand the continuum and to recognize and react to any cardiotoxicity dynamics early on for superior overall outcomes.
Collapse
Affiliation(s)
- Rhian M. Touyz
- Institute of Cardiovascular & Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
23
|
Neves KB, Rios FJ, van der Mey L, Alves-Lopes R, Cameron AC, Volpe M, Montezano AC, Savoia C, Touyz RM. VEGFR (Vascular Endothelial Growth Factor Receptor) Inhibition Induces Cardiovascular Damage via Redox-Sensitive Processes. Hypertension 2018; 71:638-647. [DOI: 10.1161/hypertensionaha.117.10490] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/07/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022]
Abstract
Although VEGF (vascular endothelial growth factor) inhibitors (VEGFIs), are effective anticancer therapies, they cause hypertension through unknown mechanisms. We questioned whether changes in vascular redox state may be important, because VEGF signaling involves nitric oxide (NO) and reactive oxygen species. Molecular mechanisms, including NOS, NADPH oxidase (Nox)–derived reactive oxygen species, antioxidant systems, and vasoconstrictor signaling pathways, were probed in human endothelial cells and vascular smooth muscle exposed to vatalanib, a VEGFI. Vascular functional effects of VEGFI were assessed ex vivo in mouse arteries. Cardiovascular and renal in vivo effects were studied in vatalanib- or gefitinib (EGFI [epidermal growth factor inhibitor])-treated mice. In endothelial cells, vatalanib decreased eNOS (Ser
1177
) phosphorylation and reduced NO and H
2
O
2
production, responses associated with increased Nox-derived O
2
−
and ONOO
−
formation. Inhibition of Nox1/4 (GKT137831) or Nox1 (NoxA1ds), prevented vatalanib-induced effects. Nrf-2 (nuclear factor erythroid 2–related factor 2) nuclear translocation and expression of Nrf-2–regulated antioxidant enzymes were variably downregulated by vatalanib. In human vascular smooth muscles, VEGFI increased Nox activity and stimulated Ca
2+
influx and MLC
20
phosphorylation. Acetylcholine-induced vasodilatation was impaired and U46619-induced vasoconstriction was enhanced by vatalanib, effects normalized by N-acetyl-cysteine and worsened by L-NAME. In vatalanib-, but not gefitinib-treated mice vasorelaxation was reduced and media:lumen ratio of mesenteric arteries was increased with associated increased cardiovascular and renal oxidative stress, decreased Nrf-2 activity and downregulation of antioxidant genes. We demonstrate that inhibition of VEGF signaling induces vascular dysfunction through redox-sensitive processes. Our findings identify Noxs and antioxidant enzymes as novel targets underling VEGFI-induced vascular dysfunction. These molecular processes may contribute to vascular toxicity and hypertension in VEGFI-treated patients.
Collapse
Affiliation(s)
- Karla B. Neves
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Francisco J. Rios
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Lucas van der Mey
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Rheure Alves-Lopes
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Alan C. Cameron
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Massimo Volpe
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Augusto C. Montezano
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Carmine Savoia
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| | - Rhian M. Touyz
- From the BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (K.B.N., F.J.R., L.v.d.M., R.A.-L., A.C.C., A.C.M., R.M.T.); Department of Clinical and Molecular Medicine, Cardiology Unit Sant’Andrea Hospital, Sapienza University of Rome, Italy (M.V., C.S.); and Department of AngioCardioNeurology and Translational Medicine, IRCCS Neuromed - Mediterranean Neurological Institute, Pozzilli, Italy (M.V.)
| |
Collapse
|
24
|
Pandey AK, Singhi EK, Arroyo JP, Ikizler TA, Gould ER, Brown J, Beckman JA, Harrison DG, Moslehi J. Mechanisms of VEGF (Vascular Endothelial Growth Factor) Inhibitor-Associated Hypertension and Vascular Disease. Hypertension 2017; 71:e1-e8. [PMID: 29279311 DOI: 10.1161/hypertensionaha.117.10271] [Citation(s) in RCA: 195] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Arvind K Pandey
- From the Division of Cardiovascular Medicine (A.K.P., E.K.S., J.B., J.A.B., J.M.), Division of Nephrology (J.P.A., T.A.I., E.R.G.), Vanderbilt Center for Kidney Disease (T.A.I.), Division of Clinical Pharmacology (D.G.H.) and Cardio-Oncology Program (J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Eric K Singhi
- From the Division of Cardiovascular Medicine (A.K.P., E.K.S., J.B., J.A.B., J.M.), Division of Nephrology (J.P.A., T.A.I., E.R.G.), Vanderbilt Center for Kidney Disease (T.A.I.), Division of Clinical Pharmacology (D.G.H.) and Cardio-Oncology Program (J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Juan Pablo Arroyo
- From the Division of Cardiovascular Medicine (A.K.P., E.K.S., J.B., J.A.B., J.M.), Division of Nephrology (J.P.A., T.A.I., E.R.G.), Vanderbilt Center for Kidney Disease (T.A.I.), Division of Clinical Pharmacology (D.G.H.) and Cardio-Oncology Program (J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Talat Alp Ikizler
- From the Division of Cardiovascular Medicine (A.K.P., E.K.S., J.B., J.A.B., J.M.), Division of Nephrology (J.P.A., T.A.I., E.R.G.), Vanderbilt Center for Kidney Disease (T.A.I.), Division of Clinical Pharmacology (D.G.H.) and Cardio-Oncology Program (J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Edward R Gould
- From the Division of Cardiovascular Medicine (A.K.P., E.K.S., J.B., J.A.B., J.M.), Division of Nephrology (J.P.A., T.A.I., E.R.G.), Vanderbilt Center for Kidney Disease (T.A.I.), Division of Clinical Pharmacology (D.G.H.) and Cardio-Oncology Program (J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Jonathan Brown
- From the Division of Cardiovascular Medicine (A.K.P., E.K.S., J.B., J.A.B., J.M.), Division of Nephrology (J.P.A., T.A.I., E.R.G.), Vanderbilt Center for Kidney Disease (T.A.I.), Division of Clinical Pharmacology (D.G.H.) and Cardio-Oncology Program (J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Joshua A Beckman
- From the Division of Cardiovascular Medicine (A.K.P., E.K.S., J.B., J.A.B., J.M.), Division of Nephrology (J.P.A., T.A.I., E.R.G.), Vanderbilt Center for Kidney Disease (T.A.I.), Division of Clinical Pharmacology (D.G.H.) and Cardio-Oncology Program (J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - David G Harrison
- From the Division of Cardiovascular Medicine (A.K.P., E.K.S., J.B., J.A.B., J.M.), Division of Nephrology (J.P.A., T.A.I., E.R.G.), Vanderbilt Center for Kidney Disease (T.A.I.), Division of Clinical Pharmacology (D.G.H.) and Cardio-Oncology Program (J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Javid Moslehi
- From the Division of Cardiovascular Medicine (A.K.P., E.K.S., J.B., J.A.B., J.M.), Division of Nephrology (J.P.A., T.A.I., E.R.G.), Vanderbilt Center for Kidney Disease (T.A.I.), Division of Clinical Pharmacology (D.G.H.) and Cardio-Oncology Program (J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
25
|
Verkaik M, van Poelgeest EM, Kwekkeboom RFJ, Ter Wee PM, van den Brom CE, Vervloet MG, Eringa EC. Myocardial contrast echocardiography in mice: technical and physiological aspects. Am J Physiol Heart Circ Physiol 2017; 314:H381-H391. [PMID: 29101165 DOI: 10.1152/ajpheart.00242.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myocardial contrast echocardiography (MCE) offers the opportunity to study myocardial perfusion defects in mice in detail. The value of MCE compared with single-photon emission computed tomography, positron emission tomography, and computed tomography consists of high spatial resolution, the possibility of quantification of blood volume, and relatively low costs. Nevertheless, a number of technical and physiological aspects should be considered to ensure reproducibility among research groups. The aim of this overview is to describe technical aspects of MCE and the physiological parameters that influence myocardial perfusion data obtained with this technique. First, technical aspects of MCE discussed in this technical review are logarithmic compression of ultrasound data by ultrasound systems, saturation of the contrast signal, and acquisition of images during different phases of the cardiac cycle. Second, physiological aspects of myocardial perfusion that are affected by the experimental design are discussed, including the anesthesia regimen, systemic cardiovascular effects of vasoactive agents used, and fluctuations in body temperature that alter myocardial perfusion. When these technical and physiological aspects of MCE are taken into account and adequately standardized, MCE is an easily accessible technique for mice that can be used to study the control of myocardial perfusion by a wide range of factors.
Collapse
Affiliation(s)
- Melissa Verkaik
- Department of Nephrology, Institute Cardiovascular Research VU, VU University Medical Centre , Amsterdam , The Netherlands.,Department of Physiology, Institute Cardiovascular Research VU, VU University Medical Centre , Amsterdam , The Netherlands
| | - Erik M van Poelgeest
- Department of Physiology, Institute Cardiovascular Research VU, VU University Medical Centre , Amsterdam , The Netherlands
| | - Rick F J Kwekkeboom
- Department of Physiology, Institute Cardiovascular Research VU, VU University Medical Centre , Amsterdam , The Netherlands
| | - Piet M Ter Wee
- Department of Nephrology, Institute Cardiovascular Research VU, VU University Medical Centre , Amsterdam , The Netherlands
| | - Charissa E van den Brom
- Department of Anaesthesiology, Institute Cardiovascular Research VU, VU University Medical Centre , Amsterdam , The Netherlands
| | - Marc G Vervloet
- Department of Nephrology, Institute Cardiovascular Research VU, VU University Medical Centre , Amsterdam , The Netherlands
| | - Etto C Eringa
- Department of Physiology, Institute Cardiovascular Research VU, VU University Medical Centre , Amsterdam , The Netherlands
| |
Collapse
|
26
|
The Role of Angiogenesis in Cancer Treatment. Biomedicines 2017. [PMID: 28635679 DOI: 10.3390/biomedicines5020034]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A number of anti-angiogenesis drugs have been FDA-approved and are being used in cancer treatment, and a number of other agents are in different stages of clinical development or in preclinical evaluation. However, pharmacologic anti-angiogenesis strategies that arrest tumor progression might not be enough to eradicate tumors. Decreased anti-angiogenesis activity in single mechanism-based anti-angiogenic strategies is due to the redundancy, multiplicity, and development of compensatory mechanism by which blood vessels are remodeled. Improving anti-angiogenesis drug efficacy will require identification of broad-spectrum anti-angiogenesis targets. These strategies may have novel features, such as increased porosity, and are the result of complex interactions among endothelial cells, extracellular matrix proteins, growth factors, pericyte, and smooth muscle cells. Thus, combinations of anti-angiogenic drugs and other anticancer strategies such as chemotherapy appear essential for optimal outcome in cancer patients. This review will focus on the role of anti-angiogenesis strategies in cancer treatment.
Collapse
|
27
|
Abstract
A number of anti-angiogenesis drugs have been FDA-approved and are being used in cancer treatment, and a number of other agents are in different stages of clinical development or in preclinical evaluation. However, pharmacologic anti-angiogenesis strategies that arrest tumor progression might not be enough to eradicate tumors. Decreased anti-angiogenesis activity in single mechanism-based anti-angiogenic strategies is due to the redundancy, multiplicity, and development of compensatory mechanism by which blood vessels are remodeled. Improving anti-angiogenesis drug efficacy will require identification of broad-spectrum anti-angiogenesis targets. These strategies may have novel features, such as increased porosity, and are the result of complex interactions among endothelial cells, extracellular matrix proteins, growth factors, pericyte, and smooth muscle cells. Thus, combinations of anti-angiogenic drugs and other anticancer strategies such as chemotherapy appear essential for optimal outcome in cancer patients. This review will focus on the role of anti-angiogenesis strategies in cancer treatment.
Collapse
Affiliation(s)
- Mehdi Rajabi
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| |
Collapse
|
28
|
Rajabi M, Mousa SA. The Role of Angiogenesis in Cancer Treatment. Biomedicines 2017; 5:E34. [PMID: 28635679 PMCID: PMC5489820 DOI: 10.3390/biomedicines5020034] [Citation(s) in RCA: 383] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/09/2017] [Accepted: 06/15/2017] [Indexed: 12/11/2022] Open
Abstract
A number of anti-angiogenesis drugs have been FDA-approved and are being used in cancer treatment, and a number of other agents are in different stages of clinical development or in preclinical evaluation. However, pharmacologic anti-angiogenesis strategies that arrest tumor progression might not be enough to eradicate tumors. Decreased anti-angiogenesis activity in single mechanism-based anti-angiogenic strategies is due to the redundancy, multiplicity, and development of compensatory mechanism by which blood vessels are remodeled. Improving anti-angiogenesis drug efficacy will require identification of broad-spectrum anti-angiogenesis targets. These strategies may have novel features, such as increased porosity, and are the result of complex interactions among endothelial cells, extracellular matrix proteins, growth factors, pericyte, and smooth muscle cells. Thus, combinations of anti-angiogenic drugs and other anticancer strategies such as chemotherapy appear essential for optimal outcome in cancer patients. This review will focus on the role of anti-angiogenesis strategies in cancer treatment.
Collapse
Affiliation(s)
- Mehdi Rajabi
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| |
Collapse
|
29
|
Li J, Sun K, Chen Z, Shi J, Zhou D, Xie G. A fluorescence biosensor for VEGF detection based on DNA assembly structure switching and isothermal amplification. Biosens Bioelectron 2017; 89:964-969. [DOI: 10.1016/j.bios.2016.09.078] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/19/2016] [Accepted: 09/22/2016] [Indexed: 10/20/2022]
|
30
|
Wasserstrum Y, Kornowski R, Raanani P, Leader A, Pasvolsky O, Iakobishvili Z. Hypertension in cancer patients treated with anti-angiogenic based regimens. CARDIO-ONCOLOGY 2015; 1:6. [PMID: 33530150 PMCID: PMC7837153 DOI: 10.1186/s40959-015-0009-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/25/2015] [Indexed: 02/08/2023]
Abstract
New anti-cancer drugs that inhibit the vascular endothelial growth factor (VEGF) signaling pathway are highly effective in the treatment of solid tumors, however concerns remain regarding their cardiovascular safety. The most common side effect of VEGF signaling pathway (VSP) inhibition is the development of systemic hypertension. We review the incidence, possible mechanisms, significance and management of hypertension in patients treated with VSP inhibitors.
Collapse
Affiliation(s)
- Yishay Wasserstrum
- Department of Cardiology, Rabin Medical Center, Petah Tikva, 49100, Israel.,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ran Kornowski
- Department of Cardiology, Rabin Medical Center, Petah Tikva, 49100, Israel.,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Pia Raanani
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel.,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avi Leader
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel.,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oren Pasvolsky
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel.,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zaza Iakobishvili
- Department of Cardiology, Rabin Medical Center, Petah Tikva, 49100, Israel. .,Sackler School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
31
|
Belcik JT, Mott BH, Xie A, Zhao Y, Kim S, Lindner NJ, Ammi A, Linden JM, Lindner JR. Augmentation of limb perfusion and reversal of tissue ischemia produced by ultrasound-mediated microbubble cavitation. Circ Cardiovasc Imaging 2015; 8:CIRCIMAGING.114.002979. [PMID: 25834183 DOI: 10.1161/circimaging.114.002979] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Ultrasound can increase tissue blood flow, in part, through the intravascular shear produced by oscillatory pressure fluctuations. We hypothesized that ultrasound-mediated increases in perfusion can be augmented by microbubble contrast agents that undergo ultrasound-mediated cavitation and sought to characterize the biological mediators. METHODS AND RESULTS Contrast ultrasound perfusion imaging of hindlimb skeletal muscle and femoral artery diameter measurement were performed in nonischemic mice after unilateral 10-minute exposure to intermittent ultrasound alone (mechanical index, 0.6 or 1.3) or ultrasound with lipid microbubbles (2×10(8) IV). Studies were also performed after inhibiting shear- or pressure-dependent vasodilator pathways, and in mice with hindlimb ischemia. Ultrasound alone produced a 2-fold increase (P<0.05) in muscle perfusion regardless of ultrasound power. Ultrasound-mediated augmentation in flow was greater with microbubbles (3- and 10-fold higher than control for mechanical index 0.6 and 1.3, respectively; P<0.05), as was femoral artery dilation. Inhibition of endothelial nitric oxide synthase attenuated flow augmentation produced by ultrasound and microbubbles by 70% (P<0.01), whereas inhibition of adenosine-A2a receptors and epoxyeicosatrienoic acids had minimal effect. Limb nitric oxide production and muscle phospho-endothelial nitric oxide synthase increased in a stepwise fashion by ultrasound and ultrasound with microbubbles. In mice with unilateral hindlimb ischemia (40%-50% reduction in flow), ultrasound (mechanical index, 1.3) with microbubbles increased perfusion by 2-fold to a degree that was greater than the control nonischemic limb. CONCLUSIONS Increases in muscle blood flow during high-power ultrasound are markedly amplified by the intravascular presence of microbubbles and can reverse tissue ischemia. These effects are most likely mediated by cavitation-related increases in shear and activation of endothelial nitric oxide synthase.
Collapse
Affiliation(s)
- J Todd Belcik
- From the Knight Cardiovascular Center, Oregon Health and Science University, Portland (J.T.B., B.H.M., A.X., Y.Z., S.K., N.J.L., A.A., J.R.L.); and La Jolla Immunology and Allergy Institute, CA (J.M.L.)
| | - Brian H Mott
- From the Knight Cardiovascular Center, Oregon Health and Science University, Portland (J.T.B., B.H.M., A.X., Y.Z., S.K., N.J.L., A.A., J.R.L.); and La Jolla Immunology and Allergy Institute, CA (J.M.L.)
| | - Aris Xie
- From the Knight Cardiovascular Center, Oregon Health and Science University, Portland (J.T.B., B.H.M., A.X., Y.Z., S.K., N.J.L., A.A., J.R.L.); and La Jolla Immunology and Allergy Institute, CA (J.M.L.)
| | - Yan Zhao
- From the Knight Cardiovascular Center, Oregon Health and Science University, Portland (J.T.B., B.H.M., A.X., Y.Z., S.K., N.J.L., A.A., J.R.L.); and La Jolla Immunology and Allergy Institute, CA (J.M.L.)
| | - Sajeevani Kim
- From the Knight Cardiovascular Center, Oregon Health and Science University, Portland (J.T.B., B.H.M., A.X., Y.Z., S.K., N.J.L., A.A., J.R.L.); and La Jolla Immunology and Allergy Institute, CA (J.M.L.)
| | - Nathan J Lindner
- From the Knight Cardiovascular Center, Oregon Health and Science University, Portland (J.T.B., B.H.M., A.X., Y.Z., S.K., N.J.L., A.A., J.R.L.); and La Jolla Immunology and Allergy Institute, CA (J.M.L.)
| | - Azzdine Ammi
- From the Knight Cardiovascular Center, Oregon Health and Science University, Portland (J.T.B., B.H.M., A.X., Y.Z., S.K., N.J.L., A.A., J.R.L.); and La Jolla Immunology and Allergy Institute, CA (J.M.L.)
| | - Joel M Linden
- From the Knight Cardiovascular Center, Oregon Health and Science University, Portland (J.T.B., B.H.M., A.X., Y.Z., S.K., N.J.L., A.A., J.R.L.); and La Jolla Immunology and Allergy Institute, CA (J.M.L.)
| | - Jonathan R Lindner
- From the Knight Cardiovascular Center, Oregon Health and Science University, Portland (J.T.B., B.H.M., A.X., Y.Z., S.K., N.J.L., A.A., J.R.L.); and La Jolla Immunology and Allergy Institute, CA (J.M.L.).
| |
Collapse
|
32
|
Lankhorst S, Saleh L, Danser AJ, van den Meiracker AH. Etiology of angiogenesis inhibition-related hypertension. Curr Opin Pharmacol 2014; 21:7-13. [PMID: 25500206 DOI: 10.1016/j.coph.2014.11.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 11/18/2014] [Accepted: 11/26/2014] [Indexed: 11/24/2022]
Abstract
Angiogenesis inhibition, targeting vascular endothelial growth factor (VEGF) or its receptors, is an established treatment for solid tumors. A common side effect of this treatment is the development of sometimes severe hypertension. This hypertension is associated with a decrease in nitric oxide production, activation of the endothelin-signaling pathway and renin suppression. The mechanism underlying activation of the endothelin-signaling pathway is not fully understood. Both activation of endothelial cells and disinhibition of the VEGF-induced suppression of endothelin production by endothelial cells may be involved. The development of hypertension can be a reason to discontinue the angiogenesis inhibitor, thereby compromising anticancer treatment, but possibly is also a biomarker for a favorable antitumor response.
Collapse
Affiliation(s)
- Stephanie Lankhorst
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Langeza Saleh
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Ah Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Anton H van den Meiracker
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
33
|
Skinner M, Philp K, Lengel D, Coverley L, Lamm Bergström E, Glaves P, Musgrove H, Prior H, Braddock M, Huby R, Curwen JO, Duffy P, Harmer AR. The contribution of VEGF signalling to fostamatinib-induced blood pressure elevation. Br J Pharmacol 2014; 171:2308-20. [PMID: 24329544 DOI: 10.1111/bph.12559] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 12/06/2013] [Accepted: 12/11/2013] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE Fostamatinib is an inhibitor of spleen tyrosine kinase (TK). In patients, fostamatinib treatment was associated with increased BP. Some TK inhibitors cause BP elevation, by inhibiting the VEGF receptor 2 (VEGFR2). Here, we have assessed the mechanistic link between fostamatinib-induced BP elevation and inhibition of VEGF signalling. EXPERIMENTAL APPROACH We used conscious rats with automated blood sampling and radio telemetry and anaesthetized rats to measure cardiovascular changes. Rat isolated aorta and isolated hearts, and human resistance vessels in vitro were also used. NO production by human microvascular endothelial cells was measured with the NO-dependent probe, DAF-FM and VEGFR2 phosphorylation was determined in mouse lung, ex vivo. KEY RESULTS In conscious rats, fostamatinib dose-dependently increased BP. The time course of the BP effect correlated closely with the plasma concentrations of R406 (the active metabolite of fostamatinib). In anaesthetized rats, infusion of R406 increased BP and decreased femoral arterial conductance. Endothelial function was unaffected, as infusion of R406 did not inhibit hyperaemia- or ACh-induced vasodilatation in rats. R406 did not affect contraction of isolated blood vessels. R406 inhibited VEGF-stimulated NO production from human endothelial cells in vitro, and treatment with R406 inhibited VEGFR2 phosphorylation in vivo. R406 inhibited VEGF-induced hypotension in anaesthetized rats. CONCLUSIONS AND IMPLICATIONS Increased vascular resistance, secondary to reduced VEGF-induced NO release from endothelium, may contribute to BP increases observed with fostamatanib. This is consistent with the elevated BP induced by other drugs inhibiting VEGF signalling, although the contribution of other mechanisms cannot be excluded.
Collapse
|
34
|
Small HY, Montezano AC, Rios FJ, Savoia C, Touyz RM. Hypertension due to antiangiogenic cancer therapy with vascular endothelial growth factor inhibitors: understanding and managing a new syndrome. Can J Cardiol 2014; 30:534-43. [PMID: 24786444 DOI: 10.1016/j.cjca.2014.02.011] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 02/20/2014] [Accepted: 02/20/2014] [Indexed: 12/21/2022] Open
Abstract
Novel antiangiogenic cancer therapies, particularly agents that block vascular endothelial growth factor (VEGF) signalling, have improved outcomes in patients with cancers and are now used as first-line therapies for some tumours. However, with VEGF inhibitors (VEGFIs) are new complications, particularly hypertension. VEGFI-induced hypertension is a dose-dependent phenomenon due to on-target effects rather than off-target effects. Increased blood pressure occurs in almost 100% of patients who take VEGFIs, with a subset who develop severe hypertension. Molecular mechanisms underlying VEGFI-induced hypertension are unclear, but endothelial dysfunction and increased vascular resistance, due to impaired nitric oxide signalling, reduced prostacyclin production, endothelin-1 (ET-1) upregulation, oxidative stress, and rarefaction have been implicated. Treatment of hypertension should be aimed at reducing the risk of short-term morbidity associated with hypertension while maintaining effective dosing of antiangiogenic therapy for optimal cancer treatment. Although specific guidelines are not yet available for the management of VEGFI-induced hypertension, angiotensin-converting enzyme inhibitors and dihydropyridine calcium channel blockers are commonly used. Severe hypertension might require reduction of VEGFI dosing, or in some cases, interruption of treatment. As more potent VEGFIs are developed and as more cancer patients are treated with VEGFIs, the burden of hypertension toxicity will increase. This will be further compounded as the use of antiangiogenic drugs broadens to include older patients and those with pre-existing cardiovascular disease. Here we focus on VEGF as a target for antiangiogenesis and how this affects increased blood pressure. Putative mechanisms underlying VEGFI-induced hypertension are highlighted and therapeutic strategies to manage such hypertension are discussed.
Collapse
Affiliation(s)
- Heather Yvonne Small
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, UK
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, UK
| | - Francisco J Rios
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, UK
| | | | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, UK.
| |
Collapse
|
35
|
Ardelean DS, Jerkic M, Yin M, Peter M, Ngan B, Kerbel RS, Foster FS, Letarte M. Endoglin and activin receptor-like kinase 1 heterozygous mice have a distinct pulmonary and hepatic angiogenic profile and response to anti-VEGF treatment. Angiogenesis 2013; 17:129-46. [PMID: 24061911 DOI: 10.1007/s10456-013-9383-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 09/09/2013] [Indexed: 02/08/2023]
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a vascular dysplasia associated with dysregulated angiogenesis and arteriovascular malformations. The disease is caused by mutations in endoglin (ENG; HHT1) or activin receptor-like kinase 1 (ALK1; HHT2) genes, coding for transforming growth factor β (TGF-β) superfamily receptors. Vascular endothelial growth factor (VEGF) has been implicated in HHT and beneficial effects of anti-VEGF treatment were recently reported in HHT patients. To investigate the systemic angiogenic phenotype of Endoglin and Alk1 mutant mice and their response to anti-VEGF therapy, we assessed microvessel density (MVD) in multiple organs after treatment with an antibody to mouse VEGF or vehicle. Lungs were the only organ showing an angiogenic defect, with reduced peripheral MVD and secondary right ventricular hypertrophy (RVH), yet distinctly associated with a fourfold increase in thrombospondin-1 (TSP-1) in Eng (+/-) versus a rise in angiopoietin-2 (Ang-2) in Alk1 (+/-) mice. Anti-VEGF treatment did reduce lung VEGF levels but interestingly, led to an increase in peripheral pulmonary MVD and attenuation of RVH; it also normalized TSP-1 and Ang-2 expression. Hepatic MVD, unaffected in mutant mice, was reduced by anti-VEGF therapy in heterozygous and wild type mice, indicating a liver-specific effect of treatment. Contrast-enhanced micro-ultrasound demonstrated a reduction in hepatic microvascular perfusion after anti-VEGF treatment only in Eng (+/-) mice. Our findings indicate that the mechanisms responsible for the angiogenic imbalance and the response to anti-VEGF therapy differ between Eng and Alk1 heterozygous mice and raise the need for systemic monitoring of anti-angiogenic therapy effects in HHT patients.
Collapse
MESH Headings
- Activin Receptors, Type I/genetics
- Activin Receptors, Type I/metabolism
- Activin Receptors, Type II
- Animals
- Antibodies, Monoclonal/pharmacology
- Endoglin
- Heterozygote
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Liver/blood supply
- Liver/metabolism
- Liver/pathology
- Lung/blood supply
- Lung/metabolism
- Lung/pathology
- Mice
- Mice, Mutant Strains
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Ribonuclease, Pancreatic/genetics
- Ribonuclease, Pancreatic/metabolism
- Telangiectasia, Hereditary Hemorrhagic/drug therapy
- Telangiectasia, Hereditary Hemorrhagic/genetics
- Telangiectasia, Hereditary Hemorrhagic/metabolism
- Telangiectasia, Hereditary Hemorrhagic/pathology
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Daniela S Ardelean
- Molecular Structure and Function Program, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Extensive tracheal necrosis after treatment of anaplastic thyroid cancer with vascular endothelial growth factor inhibitors. Ann Thorac Surg 2013; 95:2181. [PMID: 23706447 DOI: 10.1016/j.athoracsur.2012.09.081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 09/20/2012] [Accepted: 09/28/2012] [Indexed: 11/20/2022]
|
37
|
Yu W, Bai Y, Han N, Wang F, Zhao M, Huang L, Li X. Inhibition of pathological retinal neovascularization by semaphorin 3A. Mol Vis 2013; 19:1397-405. [PMID: 23825919 PMCID: PMC3695760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 06/25/2013] [Indexed: 11/01/2022] Open
Abstract
OBJECTIVE Pathological retinal angiogenesis is a major cause of vision loss. Semaphorin 3A (Sema3A), a chemorepellent guidance protein, plays crucial roles in neural and vascular patterning. To identify its role in retinal neovascularization, we investigated its antiangiogenic effects. METHODS Human umbilical vein endothelial cells (HUVECs) were used for the in vitro study, and an oxygen-induced retinopathy (OIR) mouse model was used for the in vivo study. The HUVECs were incubated with Sema3A, and cell proliferation, migration, apoptosis, cell cycle, tube formation, and c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinases (p38 MAPK) signaling pathways were measured using Cell Counting Kit-8, Transwell, flow cytometry, Matrigel assays, and western blot. C57BL/6J mouse pups were exposed to 75% oxygen for 5 days and then brought to room air and injected with Sema3A intravitreously. At postnatal day 18, the retinal nonperfused areas were measured. The in vitro and in vivo vascular endothelial growth factor-165 (VEGF165) secretion was measured using enzyme-linked immunosorbent assay. RESULTS Sema3A not only inhibited VEGF165-induced proliferation, but also induced cell cycle arrest in a dose-dependent manner. Furthermore, Sema3A inhibited migration and tube formation, both in general and in VEGF165-containing culture medium. Using an enzyme-linked immunosorbent assay, we showed that Sema3A did not affect VEGF165 secretion, but it did impede VEGF165 function. Additionally, Sema3A significantly inhibited the phosphorylation of the JNK and p38MAPK signaling pathways. When administered intravitreously, Sema3A reduced the pathological vascular changes seen in the retinal neovascularization OIR model. CONCLUSIONS These results suggest that the administration of Sema3A could be a useful therapeutic strategy for preventing hypoxia/ischemic-induced retinal neovascularization.
Collapse
Affiliation(s)
- Wenzhen Yu
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
| | - Yujing Bai
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
| | - Na Han
- Department of Orthopaedics and Trauma, Peking University People's Hospital, Beijing, China
| | - Fei Wang
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
| | - Min Zhao
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
| | - Lvzhen Huang
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
| | - Xiaoxin Li
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Department of Ophthalmology, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
38
|
Conti E, Zezza L, Ralli E, Caserta D, Musumeci MB, Moscarini M, Autore C, Volpe M. Growth factors in preeclampsia: a vascular disease model. A failed vasodilation and angiogenic challenge from pregnancy onwards? Cytokine Growth Factor Rev 2013; 24:411-25. [PMID: 23800655 DOI: 10.1016/j.cytogfr.2013.05.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 04/30/2013] [Accepted: 05/21/2013] [Indexed: 12/25/2022]
Abstract
Preeclampsia is the major cause of maternofetal and neonatal morbi-mortality including intrauterine growth retardation, miscarriages and stillbirths. Inadequate vascular dilation and angiogenesis represent the crucial underlying defect of gravidic hypertension, denoting a failed response to the vasodilatory and pro-angiogenic challenge imposed by pregnancy, especially if multifetal. A similar pathogenesis appears involved in gestational diabetes. In this review we aimed to provide a hint on understanding the deeply involved angiogenic disorders which eventually culminate in utero-placental failure. The key players in these complex processes may be found in an intricate network of growth factors (GFs) and GF inhibitors, controlled by several vascular risk factors modulated by environment and genes, which eventually impact on early and late cardiovascular outcomes of mother and fetus.
Collapse
Affiliation(s)
- E Conti
- Cardiology, Clinical and Molecular Medicine Department, "Sapienza" University of Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Bonsignore A, Warburton D. The mechanisms responsible for exercise intolerance in early-stage breast cancer: What role does chemotherapy play? Hong Kong Physiother J 2013. [DOI: 10.1016/j.hkpj.2013.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
40
|
Plate KH, Scholz A, Dumont DJ. Tumor angiogenesis and anti-angiogenic therapy in malignant gliomas revisited. Acta Neuropathol 2012; 124:763-75. [PMID: 23143192 PMCID: PMC3508273 DOI: 10.1007/s00401-012-1066-5] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 11/02/2012] [Accepted: 11/02/2012] [Indexed: 12/13/2022]
Abstract
The cellular and molecular mechanisms of tumor angiogenesis and its prospects for anti-angiogenic cancer therapy are major issues in almost all current concepts of both cancer biology and targeted cancer therapy. Currently, (1) sprouting angiogenesis, (2) vascular co-option, (3) vascular intussusception, (4) vasculogenic mimicry, (5) bone marrow-derived vasculogenesis, (6) cancer stem-like cell-derived vasculogenesis and (7) myeloid cell-driven angiogenesis are all considered to contribute to tumor angiogenesis. Many of these processes have been described in developmental angiogenesis; however, the relative contribution and relevance of these in human brain cancer remain unclear. Preclinical tumor models support a role for sprouting angiogenesis, vascular co-option and myeloid cell-derived angiogenesis in glioma vascularization, whereas a role for the other four mechanisms remains controversial and rather enigmatic. The anti-angiogenesis drug Avastin (Bevacizumab), which targets VEGF, has become one of the most popular cancer drugs in the world. Anti-angiogenic therapy may lead to vascular normalization and as such facilitate conventional cytotoxic chemotherapy. However, preclinical and clinical studies suggest that anti-VEGF therapy using bevacizumab may also lead to a pro-migratory phenotype in therapy resistant glioblastomas and thus actively promote tumor invasion and recurrent tumor growth. This review focusses on (1) mechanisms of tumor angiogenesis in human malignant glioma that are of particular relevance for targeted therapy and (2) controversial issues in tumor angiogenesis such as cancer stem-like cell-derived vasculogenesis and bone-marrow-derived vasculogenesis.
Collapse
Affiliation(s)
- Karl H Plate
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School, Frankfurt, Germany.
| | | | | |
Collapse
|
41
|
Simons M, Eichmann A. "On-target" cardiac effects of anticancer drugs: lessons from new biology. J Am Coll Cardiol 2012; 60:626-7. [PMID: 22703925 DOI: 10.1016/j.jacc.2012.01.069] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 01/09/2012] [Indexed: 11/19/2022]
|