1
|
Redfern WS, Pollard CE, Holbrook M, Islam B, Abbasi M, Mahmud J, Lambert K, Haslam A, Jo H, Khalidi H, Bielecka Z, Starkey J, Ellinger T, Bryan S, Savas A, Andrews S, Aspbury R, Rosenbrier Ribeiro L, Henderson Park KA, Vargas HM, Gilmer CR. Predicting clinical outcomes from off-target receptor interactions using Secondary Intelligence™. J Pharmacol Toxicol Methods 2025; 131:107570. [PMID: 39577752 DOI: 10.1016/j.vascn.2024.107570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Adverse effects due to off-target activity can be predicted by careful comparison of the relationship between expected plasma concentration and off-target activity of the test compound with that of reference drugs targeting that receptor for their therapeutic efficacy. The ratio between plasma concentration (unbound) and the Ki at the receptor is a surrogate measure reflecting receptor occupancy. Where data are available for reference drugs, we have curated and evaluated this at 100 receptors, 72 of which can involve both negative and positive modulations by drugs: a total of 172 'receptor modulations'. This provides a quantitative framework upon which to achieve consistent risk assessment of off-target interactions across receptors, across compounds and between assessors. It therefore represents a significant departure from an opinion-based to an evidence-based approach to secondary pharmacology. Demonstration of proof-of-principle was achieved for one of the receptor interactions (α1A-adrenoceptor antagonism leading to postural hypotension in clinical use) due to the availability of high-quality off-target Ki data for >30 drugs at this receptor.
Collapse
Affiliation(s)
- Will S Redfern
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom.
| | - Chris E Pollard
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Mark Holbrook
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Barira Islam
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Mitra Abbasi
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Joanne Mahmud
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Katie Lambert
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Augustus Haslam
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Heeseung Jo
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Hiba Khalidi
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Zofia Bielecka
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom; Pharmacoepidemiology and Pharmacoeconomics Unit, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Josh Starkey
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Thomas Ellinger
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Simon Bryan
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Angeli Savas
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Steve Andrews
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Rob Aspbury
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | | | | | - Hugo M Vargas
- Translational Safety Research, TS&BA, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Clare R Gilmer
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| |
Collapse
|
2
|
Pannucci P, Van Daele M, Cooper SL, Wragg ES, March J, Groenen M, Hill SJ, Woolard J. Role of endothelin ET A receptors in the hypertension induced by the VEGFR-2 kinase inhibitors axitinib and lenvatinib in conscious freely-moving rats. Biochem Pharmacol 2024; 228:116007. [PMID: 38145828 DOI: 10.1016/j.bcp.2023.116007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
Receptor tyrosine kinase inhibitors (RTKIs) suppress tumour growth by targeting vascular endothelial growth factor receptor 2 (VEGFR-2) which is an important mediator of angiogenesis. Here, we demonstrate that two potent RTKIs, axitinib and lenvatinib, are associated with hypertensive side effects. Doppler flowmetry was used to evaluate regional haemodynamic profiles of axitinib and lenvatinib. Male Sprague Dawley rats (350-500 g) were instrumented with Doppler flow probes (renal and mesenteric arteries and descending abdominal aorta) and catheters (jugular vein and distal abdominal aorta, via the caudal artery). Rats were dosed daily with axitinib (3 or 6 mg.kg-1) or lenvatinib (1 or 3 mg.kg-1) and regional haemodynamics were recorded over a maximum of 4 days. Both RTKIs caused significant (p < 0.05) increases in mean arterial pressure (MAP), which was accompanied by significant (p < 0.05) vasoconstriction in both the mesenteric and hindquarters vascular beds. To gain insight into the involvement of endothelin-1 (ET-1) in RTKI-mediated hypertension, we also monitored heart rate (HR) and MAP in response to axitinib or lenvatinib in animals treated with the ETA receptor selective antagonist sitaxentan (5 mg.kg-1) or the mixed ETA/ETB receptor antagonist bosentan (15 mg.kg-1) over two days. Co-treatment with bosentan or sitaxentan markedly reduced the MAP effects mediated by both RTKIs (p < 0.05). Bosentan, but not sitaxentan, also attenuated ET-1 mediated increases in HR. These data suggest that selective antagonists of ETA receptors may be appropriate to alleviate the hypertensive effects of axitinib and lenvatinib.
Collapse
Affiliation(s)
- Patrizia Pannucci
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
| | - Marieke Van Daele
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
| | - Samantha L Cooper
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
| | - Edward S Wragg
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
| | - Julie March
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Marleen Groenen
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK.
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK.
| |
Collapse
|
3
|
Qian YW, Guo YQ, Li YL, Wang Y, Guo S, Niu QQ, Zhu ML, Li P. The antihypertensive effect of Alizarin is achieved by activating VEGFR2/eNOS pathway, attenuating oxidative stress-induced mitochondrial damage and premature senescence. Life Sci 2024; 351:122862. [PMID: 38917872 DOI: 10.1016/j.lfs.2024.122862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/10/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
The primary and initial manifestations of hypertension encompass arterial hypoelasticity and histiocyte senescence. Oxidative stress plays a pivotal role in the progression of senescence. Elevated intracellular oxidative stress levels will directly induce cell damage, disrupt normal physiological signal transduction, which can cause mitochondrial dysfunction to accelerate the process of senescence. Alizarin, an anthraquinone active ingredient isolated from Rubia cordifolia L., has a variety of pharmacological effects, including antioxidant, anti-inflammatory and anti-platelet. Nevertheless, its potential in lowering blood pressure (BP) and mitigating hypertension-induced vascular senescence remains uncertain. In this study, we used spontaneously hypertensive rats (SHR) and human umbilical vein endothelial cells (HUVECs) to establish a model of vascular senescence in hypertension. Our aim was to elucidate the mechanisms underpinning the vascular protective effects of Alizarin. By assessing systolic blood pressure (SBP) and diastolic blood pressure (DBP), H&E staining, SA-β-Gal staining, vascular function, oxidative stress levels, calcium ion concentration and mitochondrial membrane potential, we found that Alizarin not only restored SBP and increased endothelium-dependent relaxation (EDR) in SHR, but also inhibited oxidative stress-induced mitochondrial damage and significantly delayed the vascular senescence effect in hypertension, and the mechanism may be related to the activation of VEGFR2/eNOS signaling pathway.
Collapse
Affiliation(s)
- Yi-Wen Qian
- Department of Pharmacy, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471000, China
| | - Ya-Qi Guo
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Yin-Lan Li
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Heilongjiang 150040, China
| | - Yang Wang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, China
| | - Qian-Qian Niu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Mo-Li Zhu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China.
| | - Peng Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, College of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China; Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, China.
| |
Collapse
|
4
|
Van Daele M, Kilpatrick LE, Woolard J, Hill SJ. Characterisation of tyrosine kinase inhibitor-receptor interactions at VEGFR2 using sunitinib-red and nanoBRET. Biochem Pharmacol 2023:115672. [PMID: 37406966 DOI: 10.1016/j.bcp.2023.115672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/13/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023]
Abstract
Vascular endothelial growth factor (VEGF) is an important mediator of angiogenesis, proliferation and migration of vascular endothelial cells. It is well known that cardiovascular safety liability for a wide range of small molecule tyrosine kinase inhibitors (TKIs) can result from interference with the VEGFR2 signalling system. In this study we have developed a ligand-binding assay using a fluorescent analogue of sunitinib (sunitinib-red) and full length VEGFR2 tagged on its C-terminus with the bioluminescent protein nanoluciferase to monitor ligand-binding to VEGFR2 using bioluminescence resonance energy transfer (BRET). This NanoBRET assay is a proximity-based assay (requiring the fluorescent and bioluminescent components to be within 10nm of each other) that can monitor the binding of ligands to the kinase domain of VEGFR2. Sunitinib-red was not membrane permeable but was able to monitor the binding affinity and kinetics of a range of TKIs in cell lysates. Kinetic studies showed that sunitinib-red bound rapidly to VEGFR2 at 25 °C and that cediranib had slower binding kinetics with an average residence time of 112 min. Comparison between the log Ki values for inhibition of binding of sunitinib-red and log IC50 values for attenuation of VEGF165a-stimulated NFAT responses showed very similar values for compounds that inhibited sunitinib-red binding. However, two compounds that failed to inhibit sunitinib-red binding (dasatinib and entospletinib) were still able to attenuate VEGFR2-mediated NFAT signalling through inhibition of downstream signalling events. These results suggest that these compounds may still exhibit cardiovascular liabilities as a result of interference with downstream VEGFR2 signalling.
Collapse
Affiliation(s)
- Marieke Van Daele
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
| | - Laura E Kilpatrick
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK; Division of Bimolecular Science and Medicinal Chemistry, School of Pharmacy, Biodiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK.
| |
Collapse
|
5
|
Fu Y, Saxu R, Ridwan KA, Yao J, Chen X, Xu X, Zheng W, Yu P, Teng Y. Losartan Alleviates the Side Effects and Maintains the Anticancer Activity of Axitinib. Molecules 2022; 27:2764. [PMID: 35566115 PMCID: PMC9101101 DOI: 10.3390/molecules27092764] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 11/17/2022] Open
Abstract
Axitinib is one of the most potent inhibitors of the vascular endothelial growth factor (VEGF) receptor and shows strong antitumor activity toward various malignant tumors. However, its severe side effects affect the quality of life and prognosis of patients. Losartan, which functions as a typical angiotensin receptor blocker, controls the average arterial pressure of patients with essential hypertension and protects against hypertension-related secondary diseases, including proteinuria and cardiovascular injury. To explore the effects of losartan on side effects caused by axitinib and its antitumor activity, several animal experiments were conducted. This study first analyzed and explored the effect of losartan on the amelioration of side effects in Wistar rats caused by axitinib. The results showed that the systolic blood pressure of Wistar rats was significantly increased by about 30 mmHg in 7 days of axitinib treatment, while the combination of losartan significantly reduced the blood pressure rise caused by axitinib. The Miles experimental model and mouse xenograft tumor model were further used to evaluate the effect of losartan on the antitumor effect of axitinib. The result clearly demonstrated that losartan has no significant influence on axitinib-related low vascular permeability and antitumor activity. In summary, our results showed that the combination of axitinib and losartan significantly reduced the side effects and maintained the antitumor effects of axitinib. This study provides information for overcoming VEGF receptor inhibitor-related side effects.
Collapse
Affiliation(s)
- Ying Fu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Rengui Saxu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Kadir Ahmad Ridwan
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Jiaping Yao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Xiaoxuan Chen
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Xueping Xu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Weida Zheng
- Medical College, Yanbian University, No. 977 Gongyuan Road, Yanji 133002, China;
| | - Peng Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| | - Yuou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Bioengineering, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.F.); (R.S.); (K.A.R.); (J.Y.); (X.C.); (X.X.)
| |
Collapse
|
6
|
Wen HN, Wang CY, Li JM, Jiao Z. Precision Cardio-Oncology: Use of Mechanistic Pharmacokinetic and Pharmacodynamic Modeling to Predict Cardiotoxicities of Anti-Cancer Drugs. Front Oncol 2022; 11:814699. [PMID: 35083161 PMCID: PMC8784755 DOI: 10.3389/fonc.2021.814699] [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: 11/14/2021] [Accepted: 12/15/2021] [Indexed: 12/18/2022] Open
Abstract
The cardiotoxicity of anti-cancer drugs presents as a challenge to both clinicians and patients. Significant advances in cancer treatments have improved patient survival rates, but have also led to the chronic effects of anti-cancer therapies becoming more prominent. Additionally, it is difficult to clinically predict the occurrence of cardiovascular toxicities given that they can be transient or irreversible, with large between-subject variabilities. Further, cardiotoxicities present a range of different symptoms and pathophysiological mechanisms. These notwithstanding, mechanistic pharmacokinetic (PK) and pharmacodynamic (PD) modeling offers an important approach to predict cardiotoxicities and offering precise cardio-oncological care. Efforts have been made to integrate the structures of physiological and pharmacological networks into PK-PD modeling to the end of predicting cardiotoxicities based on clinical evaluation as well as individual variabilities, such as protein expression, and physiological changes under different disease states. Thus, this review aims to report recent progress in the use of PK-PD modeling to predict cardiovascular toxicities, as well as its application in anti-cancer therapies.
Collapse
Affiliation(s)
- Hai-Ni Wen
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chen-Yu Wang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jin-Meng Li
- Department of Pharmacy, Affiliated Hangzhou Chest Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
Khouri C, Mahé J, Caquelin L, Locher C, Despas F. Pharmacology and pharmacovigilance of protein kinase inhibitors. Therapie 2021; 77:207-217. [PMID: 34895753 DOI: 10.1016/j.therap.2021.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/31/2021] [Indexed: 11/19/2022]
Abstract
Protein kinase inhibitors experienced their advent in the 2000s. Their market introduction made it possible to constitute a class of targeted therapies administered orally. This name was chosen to mark a break with conventional chemotherapy drugs, but it is important to stress that these are multi-target drugs with complex affinity profiles. Adverse effects can be explained by direct interactions with their targets of interest, chosen for their indications (on-target) but also interactions with other targets (off-target). The adverse effect profiles of these drugs are therefore varied and it is possible to identify common profiles related to inhibitions of common targets. Identification of these targets has improved the global understanding of the pathophysiological mechanisms underlying the onset of adverse drug reactions as well as of the related diseases, and makes it possible to predict the adverse effect profile of new protein kinase inhibitors based on their affinities. In this review, we describe the main adverse drug reactions associated with protein kinase inhibitors, their frequency and their plausible mechanisms of action.
Collapse
Affiliation(s)
- Charles Khouri
- Pharmacovigilance Department, Grenoble Alpes University Hospital, 38000 Grenoble, France; Inserm UMR 1300-HP2 Laboratory, University Grenoble Alpes, 38000 Grenoble, France
| | - Julien Mahé
- Department of Pharmacology, Regional Pharmacovigilance Center, CHU de Nantes, 44093 Nantes, France
| | - Laura Caquelin
- Inserm, CIC 1414 (centre d'investigation clinique de Rennes), Université Rennes, CHU de Rennes, 35000 Rennes, France
| | - Clara Locher
- Inserm, CIC 1414 (centre d'investigation clinique de Rennes), Université Rennes, CHU de Rennes, 35000 Rennes, France
| | - Fabien Despas
- Inserm 1297, CIC 1436, Department of Medical and Clinical Pharmacology, Faculty of Medicine, CHU de Toulouse, University Paul-Sabatier, 31000 Toulouse, France.
| |
Collapse
|
8
|
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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022]
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
|
9
|
Banks E, Grondine M, Bhavsar D, Barry E, Kettle JG, Reddy VP, Brown C, Wang H, Mettetal JT, Collins T, Adeyemi O, Overman R, Lawson D, Harmer AR, Reimer C, Drew L, Packer MJ, Cosulich S, Jones RDO, Shao W, Wilson D, Guichard S, Fawell S, Anjum R. Discovery and pharmacological characterization of AZD3229, a potent KIT/PDGFRα inhibitor for treatment of gastrointestinal stromal tumors. Sci Transl Med 2021; 12:12/541/eaaz2481. [PMID: 32350132 DOI: 10.1126/scitranslmed.aaz2481] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 03/23/2020] [Indexed: 12/12/2022]
Abstract
Gastrointestinal stromal tumor (GIST) is the most common human sarcoma driven by mutations in KIT or platelet-derived growth factor α (PDGFRα). Although first-line treatment, imatinib, has revolutionized GIST treatment, drug resistance due to acquisition of secondary KIT/PDGFRα mutations develops in a majority of patients. Second- and third-line treatments, sunitinib and regorafenib, lack activity against a plethora of mutations in KIT/PDGFRα in GIST, with median time to disease progression of 4 to 6 months and inhibition of vascular endothelial growth factor receptor 2 (VEGFR2) causing high-grade hypertension. Patients with GIST have an unmet need for a well-tolerated drug that robustly inhibits a range of KIT/PDGFRα mutations. Here, we report the discovery and pharmacological characterization of AZD3229, a potent and selective small-molecule inhibitor of KIT and PDGFRα designed to inhibit a broad range of primary and imatinib-resistant secondary mutations seen in GIST. In engineered and GIST-derived cell lines, AZD3229 is 15 to 60 times more potent than imatinib in inhibiting KIT primary mutations and has low nanomolar activity against a wide spectrum of secondary mutations. AZD3229 causes durable inhibition of KIT signaling in patient-derived xenograft (PDX) models of GIST, leading to tumor regressions at doses that showed no changes in arterial blood pressure (BP) in rat telemetry studies. AZD3229 has a superior potency and selectivity profile to standard of care (SoC) agents-imatinib, sunitinib, and regorafenib, as well as investigational agents, avapritinib (BLU-285) and ripretinib (DCC-2618). AZD3229 has the potential to be a best-in-class inhibitor for clinically relevant KIT/PDGFRα mutations in GIST.
Collapse
Affiliation(s)
- Erica Banks
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Michael Grondine
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Deepa Bhavsar
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Evan Barry
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Jason G Kettle
- Chemistry, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, UK
| | | | - Crystal Brown
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Haiyun Wang
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Jerome T Mettetal
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Teresa Collins
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, UK
| | - Oladipupo Adeyemi
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, UK
| | - Ross Overman
- Discovery Sciences, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, UK
| | - Deborah Lawson
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Alexander R Harmer
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, UK
| | - Corinne Reimer
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Lisa Drew
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | | | | | - Rhys DO Jones
- DMPK, Oncology R&D, AstraZeneca, Cambridge, CB10 1XL, UK
| | - Wenlin Shao
- Projects, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - David Wilson
- Chemistry, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, UK
| | - Sylvie Guichard
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Stephen Fawell
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA
| | - Rana Anjum
- Bioscience, Oncology R&D, AstraZeneca, 35 Gatehouse Park, Boston, MA 02451, USA.
| |
Collapse
|
10
|
Design and optimisation of dendrimer-conjugated Bcl-2/x L inhibitor, AZD0466, with improved therapeutic index for cancer therapy. Commun Biol 2021; 4:112. [PMID: 33495510 PMCID: PMC7835349 DOI: 10.1038/s42003-020-01631-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 12/21/2020] [Indexed: 02/08/2023] Open
Abstract
Dual Bcl-2/Bcl-xL inhibitors are expected to deliver therapeutic benefit in many haematological and solid malignancies, however, their use is limited by tolerability issues. AZD4320, a potent dual Bcl-2/Bcl-xL inhibitor, has shown good efficacy however had dose limiting cardiovascular toxicity in preclinical species, coupled with challenging physicochemical properties, which prevented its clinical development. Here, we describe the design and development of AZD0466, a drug-dendrimer conjugate, where AZD4320 is chemically conjugated to a PEGylated poly-lysine dendrimer. Mathematical modelling was employed to determine the optimal release rate of the drug from the dendrimer for maximal therapeutic index in terms of preclinical anti-tumour efficacy and cardiovascular tolerability. The optimised candidate is shown to be efficacious and better tolerated in preclinical models compared with AZD4320 alone. The AZD4320-dendrimer conjugate (AZD0466) identified, through mathematical modelling, has resulted in an improved therapeutic index and thus enabled progression of this promising dual Bcl-2/Bcl-xL inhibitor into clinical development. Claire Patterson et al. present the design and development of AZD0466, a drug-dendrimer conjugate, and use preclinical and mathematical models to determine the optimal release rate of the drug from the dendrimer carrier for maximal therapeutic index in terms of anti-tumour efficacy and cardiovascular tolerability. This study identifies this promising dual Bcl-2/Bcl-xL inhibitor for progression to clinical development.
Collapse
|
11
|
Versmissen J, Mirabito Colafella KM, Koolen SLW, Danser AHJ. Vascular Cardio-Oncology: Vascular Endothelial Growth Factor inhibitors and hypertension. Cardiovasc Res 2020; 115:904-914. [PMID: 30726882 DOI: 10.1093/cvr/cvz022] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/16/2018] [Accepted: 01/24/2019] [Indexed: 12/27/2022] Open
Abstract
Since the formation of new blood vessels is essential for tumour growth and metastatic spread, inhibition of angiogenesis by targeting the vascular endothelial growth factor (VEGF) pathway is an effective strategy for various types of cancer, most importantly renal cell carcinoma, thyroid cancer, and hepatocellular carcinoma. However, VEGF inhibitors have serious side effects, most importantly hypertension and nephropathy. In case of fulminant hypertension, this may only be handled by lowering the dosage since the blood pressure rise is proportional to the amount of VEGF inhibition. These effects pathophysiologically and clinically resemble the most severe complication of pregnancy, preeclampsia, in which case an insufficient placenta leads to a rise in sFlt-1 levels causing a decrease in VEGF availability. Due to this overlap, studies in preeclampsia may provide important information for VEGF inhibitor-induced toxicity and vice versa. In both VEGF inhibitor-induced toxicity and preeclampsia, endothelin (ET)-1 appears to be a pivotal player. In this review, after briefly summarizing the anticancer effects, we discuss the mechanisms that potentially underlie the unwanted effects of VEGF inhibitors, focusing on ET-1, nitric oxide and oxidative stress, the renin-angiotensin-aldosterone system, and rarefaction. Given the salt sensitivity of this phenomenon, as well as the beneficial effects of aspirin in preeclampsia and cancer, we next provide novel treatment options for VEGF inhibitor-induced toxicity, including salt restriction, ET receptor blockade, and cyclo-oxygenase inhibition, in addition to classical antihypertensive and renoprotective drugs. We conclude with the recommendation of therapeutic drug monitoring to improve patient outcome.
Collapse
Affiliation(s)
- Jorie Versmissen
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, CA Rotterdam, The Netherlands
| | - Katrina M Mirabito Colafella
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, CA Rotterdam, The Netherlands.,Cardiovascular Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Physiology, Monash University, Melbourne, Australia
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.,Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, CA Rotterdam, The Netherlands
| |
Collapse
|
12
|
Pilla Reddy V, Anjum R, Grondine M, Smith A, Bhavsar D, Barry E, Guichard SM, Shao W, Kettle JG, Brown C, Banks E, Jones RDO. The Pharmacokinetic-Pharmacodynamic (PKPD) Relationships of AZD3229, a Novel and Selective Inhibitor of KIT, in a Range of Mouse Xenograft Models of GIST. Clin Cancer Res 2020; 26:3751-3759. [PMID: 32220888 DOI: 10.1158/1078-0432.ccr-19-2848] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/15/2020] [Accepted: 03/23/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE The emergence of secondary mutations is a cause of resistance to current KIT inhibitors used in the treatment of patients with gastrointestinal stromal tumors (GIST). AZD3229 is a selective inhibitor of wild-type KIT and a wide spectrum of primary and secondary mutations seen in patients with GIST. The objective of this analysis is to establish the pharmacokinetic-pharmacodynamic (PKPD) relationship of AZD3229 in a range of mouse GIST tumor models harboring primary and secondary KIT mutations, and to benchmark AZD3229 against other KIT inhibitors. EXPERIMENTAL DESIGN A PKPD model was developed for AZD3229 linking plasma concentrations to inhibition of phosphorylated KIT using data generated from several in vivo preclinical tumor models, and in vitro data generated in a panel of Ba/F3 cell lines. RESULTS AZD3229 drives inhibition of phosphorylated KIT in an exposure-dependent manner, and optimal efficacy is observed when >90% inhibition of KIT phosphorylation is sustained over the dosing interval. Integrating the predicted human pharmacokinetics into the mouse PKPD model predicts that an oral twice daily human dose greater than 34 mg is required to ensure adequate coverage across the mutations investigated. Benchmarking shows that compared with standard-of-care KIT inhibitors, AZD3229 has the potential to deliver the required target coverage across a wider spectrum of primary or secondary mutations. CONCLUSIONS We demonstrate that AZD3229 warrants clinical investigation as a new treatment for patients with GIST based on its ability to inhibit both ATP-binding and A-loop mutations of KIT at clinically relevant exposures.
Collapse
Affiliation(s)
| | - Rana Anjum
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Michael Grondine
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Aaron Smith
- Research and Early Development, Oncology R&D, AstraZeneca, United Kingdom
| | - Deepa Bhavsar
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Evan Barry
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Sylvie M Guichard
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Wenlin Shao
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Jason G Kettle
- Research and Early Development, Oncology R&D, AstraZeneca, United Kingdom
| | - Crystal Brown
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Erica Banks
- Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Rhys D O Jones
- Research and Early Development, Oncology R&D, AstraZeneca, United Kingdom.
| |
Collapse
|
13
|
Collins TA, Rolf MG, Pointon A. Current and future approaches to nonclinical cardiovascular safety assessment. Drug Discov Today 2020; 25:1129-1134. [PMID: 32209428 DOI: 10.1016/j.drudis.2020.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/03/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022]
Abstract
Our goal is to accurately predict all types of cardiovascular events in patients utilising nonclinical cardiovascular safety data. In the past two decades, cardiovascular safety science has primarily focused on events associated with the electrocardiogram. Broadening out to other cardiovascular parameters, we share real-life case studies that highlight our progress towards improved and better-informed project progression based upon use of disease models, mechanism-based translation and structure-function relationships. To fulfil this goal, further advances in patient-relevant humanised models will be required to enable cardiovascular safety science to keep pace with the ever-changing landscape of novel therapeutic paradigms.
Collapse
Affiliation(s)
- Teresa A Collins
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Michael G Rolf
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Amy Pointon
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
14
|
Vlasov TD, Nesterovich II, Shimanski DA. Endothelial dysfunction: from the particular to the general. Return to the «Old Paradigm»? ACTA ACUST UNITED AC 2019. [DOI: 10.24884/1682-6655-2019-18-2-19-27] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The vascular endothelium is a heterogeneous structure with diverse functions, being an active metabolic system. Endothelial cells mediate inflammatory and immune processes, regulate leukocyte adhesion, permeability and vascular tone, participate in the hemostasis system, stimulate the processes of angiogenesis. Endothelial dysfunction can initiate individual disorders, but more often it is a universal link in the pathogenesis of many diseases. Currently, endothelial dysfunction is presented as an imbalance between the production of vasodilating, angioprotective, antiproliferative factors, on the one hand, and vasoconstrictive, prothrombotic, proliferative factors, on the other hand. The manifestations of endothelial dysfunction, the direction and severity of these changes may vary depending on the disease. The review provides examples of combined endothelial disorders in the most studied and common diseases (essential hypertension, type 2 diabetes, systemic diseases of the connective tissue, atherosclerosis, and malignant tumors). Despite the presence of rare cases of isolated endothelial dysfunction, it can be argued that in the absolute majority of diseases, endothelial dysfunction has combined type of violations. The allocation of individual endothelial disorder spectra, typical for a specific disease, is problematic, due to the universality and nonspecificity of the manifestations of endothelial dysfunction. These conclusions allow us to return to the origins of this problem, considering endothelial dysfunction as a holistic concept, not limited to a certain range of its disorders.
Collapse
|
15
|
Upreti VV, Venkatakrishnan K. Model‐Based Meta‐Analysis: Optimizing Research, Development, and Utilization of Therapeutics Using the Totality of Evidence. Clin Pharmacol Ther 2019; 106:981-992. [DOI: 10.1002/cpt.1462] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/21/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Vijay V. Upreti
- Clinical Pharmacology Modeling and SimulationAmgen Inc. South San Francisco California USA
| | - Karthik Venkatakrishnan
- Quantitative Clinical PharmacologyTakeda Pharmaceuticals International Co. Cambridge Massachusetts USA
| |
Collapse
|
16
|
Johansson J, Larsson MH, Hornberg JJ. Predictive in vitro toxicology screening to guide chemical design in drug discovery. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
17
|
Cooper SL, Carter JJ, March J, Woolard J. Long-term cardiovascular effects of vandetanib and pazopanib in normotensive rats. Pharmacol Res Perspect 2019; 7:e00477. [PMID: 31164986 PMCID: PMC6543457 DOI: 10.1002/prp2.477] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/08/2019] [Indexed: 01/24/2023] Open
Abstract
Vandetanib and pazopanib are clinically available, multi-targeted inhibitors of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptor tyrosine kinases. Short-term VEGF receptor inhibition is associated with hypertension in 15%-60% of patients, which may limit the use of these anticancer therapies over the longer term. To evaluate the longer-term cardiovascular implications of treatment, we investigated the "on"-treatment (21 days) and "off"-treatment (10 days) effects following daily administration of vandetanib, pazopanib, or vehicle, in conscious rats. Cardiovascular variables were monitored in unrestrained Sprague-Dawley rats instrumented with radiotelemetric devices. In Study 1, rats were randomly assigned to receive either daily intraperitoneal injections of vehicle (volume 0.5 mL; n = 5) or vandetanib 25 mg/kg/day (volume 0.5 mL; n = 6). In Study 2, rats received either vehicle (volume 0.5 mL; n = 4) or pazopanib 30 mg/kg/day (volume 0.5 mL; n = 7), dosed once every 24 hours for 21 days. All solutions were in 2% Tween, 5% propylene glycol in 0.9% saline solution. Vandetanib caused sustained increases in mean arterial pressure (MAP), systolic blood pressure (SBP), and diastolic blood pressure (DBP) compared to baseline and vehicle. Vandetanib also significantly altered the circadian cycling of MAP, SBP, and DBP. Elevations in SBP were detectable 162 hours after the last dose of vandetanib. Pazopanib also caused increases in MAP, SBP, and DBP. However, compared to vandetanib, these increases were of slower onset and a smaller magnitude. These data suggest that the cardiovascular consequences of vandetanib and pazopanib treatment are sustained, even after prolonged cessation of drug treatment.
Collapse
Affiliation(s)
- Samantha L. Cooper
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
| | - Joanne J. Carter
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
| | - Julie March
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and NeuroscienceSchool of Life SciencesQueen's Medical CentreUniversity of NottinghamNottinghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)University of Birmingham and University of NottinghamMidlandsUK
| |
Collapse
|
18
|
Skinner M, Ceuppens P, White P, Prior H. Social-housing and use of double-decker cages in rat telemetry studies. J Pharmacol Toxicol Methods 2019; 96:87-94. [DOI: 10.1016/j.vascn.2019.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/21/2019] [Accepted: 02/11/2019] [Indexed: 11/16/2022]
|
19
|
Chen Q, Yang Z, Chen X, Shu L, Qu W. Peptide P7 inhibits the bFGF-stimulated proliferation and invasion of SKOV3 cells. Exp Ther Med 2019; 17:3003-3008. [PMID: 30936970 PMCID: PMC6434263 DOI: 10.3892/etm.2019.7309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 01/31/2019] [Indexed: 12/11/2022] Open
Abstract
Peptide P7 specifically binds with basic fibroblast growth factor (bFGF) to inhibit the proliferation and invasion of numerous types of cancer cell. However, this effect has remained to be demonstrated in ovarian cancer-derived cell lines. In the present study, the protein P7 was used treat bFGF-stimulated SKOV3 epithelial ovarian cancer cells to explore the therapeutic potential of P7. An MTT and a scratch wound assay were used to respectively evaluate the proliferation and migration of bFGF-stimulated SKOV3 cells treated with P7. Reverse transcription-quantitative polymerase chain reaction analysis was used to detect the gene expression of urokinase-type plasminogen activator (uPA), as well as matrix metallopeptidase (MMP)-2 and −9, which have a role in cell migration/invasion. The morphology and proliferation of SKOV3 cells were not significantly affected by different concentrations of P7. However, P7 had an obvious inhibitory effect on the proliferation and migration of bFGF-stimulated SKOV3 cells. Treatment with P7 significantly lowered the gene expression of uPA, MMP-2 and MMP-9 compared with that in the control group. In conclusion, the present results suggested that P7, which, at least in part, acts through inhibition of bFGF, may have a potential therapeutic application in epithelial ovarian cancer.
Collapse
Affiliation(s)
- Qiong Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Ziying Yang
- Department of Obstetrics and Gynecology, Shangrao People's Hospital, Shangrao, Jiangxi 334000, P.R. China
| | - Xiangnan Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Li Shu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Wanglei Qu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
20
|
Collins T, Gray K, Bista M, Skinner M, Hardy C, Wang H, Mettetal JT, Harmer AR. Quantifying the relationship between inhibition of VEGF receptor 2, drug-induced blood pressure elevation and hypertension. Br J Pharmacol 2018; 175:618-630. [PMID: 29161763 DOI: 10.1111/bph.14103] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/20/2017] [Accepted: 11/11/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Several anti-angiogenic cancer drugs that inhibit VEGF receptor (VEGFR) signalling for efficacy are associated with a 15-60% incidence of hypertension. Tyrosine kinase inhibitors (TKIs) that have off-target activity at VEGFR-2 may also cause blood pressure elevation as an undesirable side effect. Therefore, the ability to translate VEGFR-2 off-target potency into blood pressure elevation would be useful in development of novel TKIs. Here, we have sought to quantify the relationship between VEGFR-2 inhibition and blood pressure elevation for a range of kinase inhibitors. EXPERIMENTAL APPROACH Porcine aortic endothelial cells overexpressing VEGFR-2 (PAE) were used to determine IC50 for VEGFR-2 phosphorylation. These IC50 values were compared with published reports of exposure attained during clinical use and the corresponding incidence of all-grade hypertension. Unbound average plasma concentration (Cav,u ) was selected to be the most appropriate pharmacokinetic parameter. The pharmacokinetic-pharmacodynamic (PKPD) relationship for blood pressure elevation was investigated for selected kinase inhibitors, using data derived either from clinical papers or from rat telemetry experiments. KEY RESULTS All-grade hypertension was predominantly observed when the Cav,u was >0.1-fold of the VEGFR-2 (PAE) IC50 . Furthermore, based on the PKPD analysis, an exposure-dependent blood pressure elevation >1 mmHg was observed only when the Cav,u was >0.1-fold of the VEGFR-2 (PAE) IC50 . CONCLUSIONS AND IMPLICATIONS Taken together, these data show that the risk of blood pressure elevation is proportional to the amount of VEGFR-2 inhibition, and a margin of >10-fold between VEGFR-2 IC50 and Cav,u appears to confer a minimal risk of hypertension.
Collapse
Affiliation(s)
- Teresa Collins
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Kelly Gray
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Michal Bista
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Matt Skinner
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Christopher Hardy
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Haiyun Wang
- AstraZeneca, Gatehouse Park, Waltham, MA, 02451, USA
| | | | - Alexander R Harmer
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| |
Collapse
|