1
|
Liu S, Liu C, He Y, Li J. Benign non-immune cells in tumor microenvironment. Front Immunol 2025; 16:1561577. [PMID: 40248695 PMCID: PMC12003390 DOI: 10.3389/fimmu.2025.1561577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 04/19/2025] Open
Abstract
The tumor microenvironment (TME) is a highly complex and continuous evolving ecosystem, consisting of a diverse array of cellular and non-cellular components. Among these, benign non-immune cells, including cancer-associated fibroblasts (CAFs), adipocytes, endothelial cells (ECs), pericytes (PCs), Schwann cells (SCs) and others, are crucial factors for tumor development. Benign non-immune cells within the TME interact with both tumor cells and immune cells. These interactions contribute to tumor progression through both direct contact and indirect communication. Numerous studies have highlighted the role that benign non-immune cells exert on tumor progression and potential tumor-promoting mechanisms via multiple signaling pathways and factors. However, these benign non-immune cells may play different roles across cancer types. Therefore, it is important to understand the potential roles of benign non-immune cells within the TME based on tumor heterogeneity. A deep understanding allows us to develop novel cancer therapies by targeting these cells. In this review, we will introduce several types of benign non-immune cells that exert on different cancer types according to tumor heterogeneity and their roles in the TME.
Collapse
Affiliation(s)
- Shaowen Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chunhui Liu
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Yuan He
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Li
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
2
|
Sato MT, Yashima H, Araki T, Nomura M, Suzuki K, Yamamoto K. Effect of dasatinib on blood concentrations of sunitinib and adverse events in a patient with metastatic renal cell carcinoma treated with sunitinib: A case report. Exp Ther Med 2025; 29:69. [PMID: 39991724 PMCID: PMC11843194 DOI: 10.3892/etm.2025.12819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
The combined use of sunitinib and other tyrosine kinase inhibitors (TKIs) is discouraged because of the increased risk of adverse events (AEs). Furthermore, plasma sunitinib levels are affected by drugs that affect CYP3A4 activity; therefore, caution should be exercised when using CYP3A4 inhibitors. In the present study, a 59-year-old Japanese man with metastatic renal cell carcinoma (RCC) was diagnosed with chronic myeloid leukemia (CML) while on sunitinib treatment and was simultaneously treated with sunitinib and dasatinib, a multi-TKI used for CML with moderate CYP3A4 inhibitory activity. The trough levels of sunitinib and N-desethyl sunitinib were 63.7 and 13.7 ng/ml, respectively, with sunitinib 50 mg/day alone. While grade 2 hand-foot skin reactions and grade 2 diarrhea were observed after starting dasatinib, the trough levels of sunitinib and N-desethyl sunitinib were stable, and dasatinib levels were lower than the reference range. Because of the risk of severe AEs, the doses of sunitinib and dasatinib were temporarily reduced or suspended. Ultimately, they were maintained at 87.5 and 83.3% of their initial doses, respectively, with no severe AEs observed. The patient achieved a complete cytogenetic response for CML on day 154 after starting dasatinib treatment; however, RCC metastasis was observed on day 186, leading to a switch from sunitinib to axitinib. This suggests that dasatinib did not significantly affect the plasma levels of sunitinib. A dose reduction at the start of combination therapy is advisable, increasing the dose while monitoring AEs may safely provide sufficient therapeutic intensity.
Collapse
Affiliation(s)
- Miki Takenaka Sato
- Department of Pharmacy, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
- Division of Pharmacotherapeutics, Department of Clinical Pharmacy, School of Pharmacy, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Hideaki Yashima
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Takuya Araki
- Department of Pharmacy, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Masashi Nomura
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Kazuhiro Suzuki
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Koujirou Yamamoto
- Department of Pharmacy, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
3
|
Qian J, Ruan C, Cai Y, Yi W, Liu J, Xu R. Development and validation of a risk-prediction model for adverse drug reactions in real-world cancer patients treated with anlotinib. Ther Adv Drug Saf 2025; 16:20420986251328687. [PMID: 40151494 PMCID: PMC11946284 DOI: 10.1177/20420986251328687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Background The risk factors related to the adverse drug reactions (ADRs) of anlotinib have been rarely investigated. In addition, a corresponding risk prediction model has not been established in China pertaining to anlotinib-related ADRs. Objectives To manage ADRs more efficiently and improve the prognosis of patients administered anlotinib. Design A retrospective analysis was conducted using the medical records of patients diagnosed with cancer who were administered anlotinib after hospitalization between January 1, 2020, and December 31, 2023. Methods We performed a combination of univariate analysis and multivariate binary logistic regression analysis to identify significant factors that can accurately predict ADRs. Model fitting was performed using forward selection. The accuracy of the prediction model was expressed as the area under the receiver operating characteristic curve (AUC). The final ADR risk model was validated. Results In this study, 300 patients who were administered anlotinib were included. Among them, 238 (79.33%) patients experienced at least one ADR. Diagnosis, combination treatment, distant metastasis, treatment lines, and cumulative dose were independent risk factors for the ADRs of anlotinib. The AUC and the concordance index of the nomogram constructed from the above five factors were 0.790 and 0.789, respectively. The results of the Hosmer-Lemeshow test showed that the model was a good fit (p = 0.811). In addition, the decision curve analysis demonstrated a significantly higher net benefit of the model. The external validation indicated that the prediction nomogram was reliable. Conclusion We developed and validated a simple model to use the ADR risk score in patients who were administered anlotinib. This risk prediction model was well-calibrated and discriminative. It can be used as a reference for clinical decision-making. It has clinical significance for preventing ADRs, improving the prognosis of patients, and providing support for the rational use of drugs.
Collapse
Affiliation(s)
- Jiajia Qian
- Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Cong Ruan
- Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yunyun Cai
- Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Weiyi Yi
- Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jiyong Liu
- Minhang Branch, Fudan University Shanghai Cancer Center, No. 106, Ruili Road, Shanghai 200240, China
- Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rui Xu
- Minhang Branch, Fudan University Shanghai Cancer Center, No. 106, Ruili Road, Shanghai 200240, China
| |
Collapse
|
4
|
Rekowski J, Guo C, Solovyeva O, Dimairo M, Rouhifard M, Patel D, Alger E, Ashby D, Berlin J, Boix O, Calvert M, Chan AW, Coschi CH, de Bono J, Evans TRJ, Garrett–Mayer E, Golub RM, Hayward KS, Hopewell S, Isaacs JD, Ivy SP, Jaki T, Kholmanskikh O, Kightley A, Lee S, Liu R, Maia I, Mander A, Marshall LV, Matcham J, Peck R, Rantell KR, Richards DP, Seymour L, Tanaka Y, Ursino M, Weir CJ, Yap C. CONSORT-DEFINE explanation and elaboration: recommendations for enhancing reporting quality and impact of early phase dose-finding clinical trials. EClinicalMedicine 2025; 79:102987. [PMID: 39877553 PMCID: PMC11773258 DOI: 10.1016/j.eclinm.2024.102987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 01/31/2025] Open
Abstract
Early phase dose-finding (EPDF) trials are key in the development of novel therapies, with their findings directly informing subsequent clinical development phases and providing valuable insights for reverse translation. Comprehensive and transparent reporting of these studies is critical for their accurate and critical interpretation, which may improve and expedite therapeutic development. However, quality of reporting of design characteristics and results from EPDF trials is often variable and incomplete. The international consensus-based CONSORT-DEFINE (Consolidated Standards for Reporting Trials Dose-finding Extension) statement, an extension of the CONSORT statement for randomised trials, was developed to improve the reporting of EPDF trials. The CONSORT-DEFINE statement introduced 21 new items and modified 19 existing CONSORT items.This CONSORT-DEFINE Explanation and Elaboration (E&E) document provides important information to enhance understanding and facilitate the implementation of the CONSORT-DEFINE checklist. For each new or modified checklist item, we provide a detailed description and its rationale with supporting evidence, and present examples from EPDF trial reports published in peer-reviewed scientific journals. When reporting the results of EPDF trials, authors are encouraged to consult the CONSORT-DEFINE E&E document, together with the CONSORT and CONSORT-DEFINE statement papers, and adhere to their recommendations. Widespread adoption of the CONSORT-DEFINE statement is likely to enhance the reporting quality of EPDF trials, thus facilitating the peer review of such studies and their appraisal by researchers, regulators, ethics committee members, and funders. Funding This work is a further extension of the CONSORT-DEFINE study, which was funded by the UK Medical Research Council (MRC)-National Institute for Health and Care Research (NIHR) Methodology Research Programme (MR/T044934/1). The Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU) receives programmatic infrastructure funding from Cancer Research UK (C1491/A25351; CTUQQR-Dec 22/100 004), which has contributed to accelerating the advancement and successful completion of this work.
Collapse
Affiliation(s)
- Jan Rekowski
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Christina Guo
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Olga Solovyeva
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Munyaradzi Dimairo
- Division of Population Health, Sheffield Centre for Health and Related Research, University of Sheffield, Sheffield, UK
| | - Mahtab Rouhifard
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Dhrusti Patel
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Emily Alger
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Deborah Ashby
- School of Public Health, Imperial College London, St Mary's Hospital, London, UK
| | | | | | - Melanie Calvert
- Centre for Patient Reported Outcomes Research, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Birmingham Health Partners Centre for Regulatory Science and Innovation, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Applied Research Collaboration West Midlands, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Precision Transplant and Cellular Therapeutics, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Birmingham Biomedical Research Centre, NIHR Birmingham Biomedical Research Centre, Institute of Translational Medicine, University Hospital NHS Foundation Trust, Birmingham, UK
| | - An-Wen Chan
- Department of Medicine, Women's College Research Institute, University of Toronto, Toronto, Canada
| | | | - Johann de Bono
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Thomas R. Jeffry Evans
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - Elizabeth Garrett–Mayer
- Center for Research and Analytics, American Society of Clinical Oncology, Alexandria, VA, USA
| | - Robert M. Golub
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathryn S. Hayward
- Departments of Physiotherapy and Medicine, University of Melbourne, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Sally Hopewell
- Oxford Clinical Research Unit, NDORMS, University of Oxford, Oxford, UK
| | - John D. Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Freeman Hospital, Newcastle upon Tyne, UK
| | - S. Percy Ivy
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Institute of Health, Bethesda, MD, USA
| | - Thomas Jaki
- MRC Biostatistics Unit, Cambridge University, Cambridge, UK
- Computational Statistics Group, University of Regensburg, Regensburg, Germany
| | | | - Andrew Kightley
- Patient and Public Involvement and Engagement (PPIE) Lead, Lichfield, UK
| | - Shing Lee
- Columbia University Mailman School of Public Health, New York, NY, USA
| | | | | | - Adrian Mander
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Lynley V. Marshall
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - James Matcham
- Strategic Consulting, Cytel (Australia), Perth, WA, Australia
| | - Richard Peck
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Hoffmann-La Roche, Basel, Switzerland
| | | | | | | | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Moreno Ursino
- ReCAP/F CRIN, INSERM, 5400, Nancy, France
- Unit of Clinical Epidemiology, University Hospital Centre Robert Debré, Université Paris Cité, Paris, France
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
- HeKA Team, Centre Inria, Paris, France
| | - Christopher J. Weir
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Christina Yap
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| |
Collapse
|
5
|
Hammel P, Smith D, Afchain P, Dominguez-Tinajero S, Seitz JF, Lievre A, Van Cutsem E, Assenat E, Di Fiore F, Peeters M, Sobhani I, Raymond E, Charton E, Vernerey D, De Mestier L, Lombard-Bohas C. SUNLAND: a randomized, double-blinded phase II GERCOR trial of sunitinib versus placebo and lanreotide in patients with advanced progressive midgut neuroendocrine tumors. Ther Adv Med Oncol 2024; 16:17588359241290140. [PMID: 39563716 PMCID: PMC11574894 DOI: 10.1177/17588359241290140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/23/2024] [Indexed: 11/21/2024] Open
Abstract
Background Sunitinib, a multitarget tyrosine kinase inhibitor, showed encouraging antitumor activity and manageable toxicity in patients with advanced midgut neuroendocrine tumors (NETs) in earlier results from phase I and II trials. Patients and methods In this phase II trial, patients with a nonresectable grade 1 or 2 midgut progressive NET and Eastern Cooperative Oncology Group performance status 0-1 were randomly assigned 1:1 to receive 37.5 mg sunitinib or a placebo, combined with 120 mg lanreotide autogel every 28 days. The planned sample size was 104 patients. The primary outcome was investigator-assessed progression-free survival (PFS). Results The study was stopped early because of insufficient patient recruitment. Between January 2013 and December 2016, 44 patients were enrolled and received sunitinib (n = 22) or placebo (n = 22). The median age was 63.7 years (Q1-Q3 range, 56.6-68.1) and 26 patients (59.1%) were male. The main localization was ileum (N = 37, 84.1%) and the majority were grade 2 (n = 25, 56.8%). The median follow-up was 36.7 months (95% confidence interval (CI) 34.6-48.2). The median PFS was 9.84 months (95% CI 6.8-23.3) with sunitinib and 11.47 months (95% CI 5.4-15.3) with placebo (hazard ratio (HR) = 0.80, 95% CI 0.41-1.56, p = 0.51). There was no difference in overall survival between treatment arms (HR = 0.81, (95% CI 0.32-2.01), p = 0.64). The objective response rate was 9.1% with sunitinib and 0.0% with placebo, and 19 patients (86.4%) had stable disease. Thirty-nine patients (88.6%) completed the baseline QLQ-C30 questionnaire. Baseline health-related quality of life level was similar between treatment arms, except for physical and emotional functioning which were higher (p = 0.089) and lower (p = 0.023) in the sunitinib arm, respectively. Trends toward longer time until a definitive deterioration in favor of the sunitinib arm were observed for 10 out of 15 dimensions (HRs < 1), with a significant result for financial difficulties (HR = 0.31, (90% CI 0.10-0.94)). Twenty-seven patients (61.4%) had at least one adverse event grade ⩾3 (sunitinib: 72.7%, placebo: 50.0%), with only one patient grade 4 for hypertension and vomiting. Eleven deaths non-related to treatment occurred (sunitinib arm: n = 5, placebo arm: n = 6). Conclusion Our study does not provide enough evidence to conclude the role of sunitinib in advanced midgut NETs, primarily due to a lower-than-expected number of enrolled patients. While we cannot entirely rule out the efficacy of sunitinib, lanreotide alone may play a significant role. Trial registration EudraCT: 2012-001098-94.
Collapse
Affiliation(s)
- Pascal Hammel
- Digestive and Medical Oncology Department, Hôpital Paul Brousse, University of Paris-Saclay, 12 Avenue Paul Vaillant-Couturier, 94800 Villejuif, France
| | | | - Pauline Afchain
- Hôpital Saint-Antoine (AP-HP), University of Paris, Paris, France
| | | | | | | | | | | | | | | | | | | | - Emilie Charton
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France
- Departments of Clinical Research and Innovation and Human and Social Sciences, Centre Léon Bérard, Lyon, France
| | - Dewi Vernerey
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France
| | | | | |
Collapse
|
6
|
Lu GR, Wang RZ, Zhao XY, Xu JE, Huang CK, Sun W, Chen RJ, Wang Z. The CYP3A inducer dexamethasone affects the pharmacokinetics of sunitinib by accelerating its metabolism in rats. Chem Biol Interact 2024; 403:111228. [PMID: 39244184 DOI: 10.1016/j.cbi.2024.111228] [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: 07/01/2024] [Revised: 08/13/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Sunitinib, a novel anti-tumor small molecule targeting VEGFR, is prescribed for advanced RCC and GISTs. Sunitinib is primarily metabolized by the CYP3A enzyme. It is well-known that dexamethasone serves as a potent inducer of this enzyme system. Nonetheless, the effect of dexamethasone on sunitinib metabolism remains unclear. This study examined the effect of dexamethasone on the pharmacokinetics of sunitinib and its metabolite N-desethyl sunitinib in rats. The plasma levels of both compounds were measured using UHPLC-MS/MS. Pharmacokinetic parameters and metabolite ratio values were calculated. Compare to control group, the low-dose dexamethasone group and high-dose dexamethasone group decreased the AUC(0-t) values of sunitinib by 47 % and 45 %, respectively. Meanwhile, the AUC(0-t) values of N-desethyl sunitinib were increased by 2.2-fold and 2.4-fold in low-dose dexamethasone group and high-dose dexamethasone group, respectively. The CL values for sunitinib were both approximately 45 % higher in the two dexamethasone groups. Remarkably, metabolite ratio values increased over 5-fold in both low-dose dexamethasone group and high-dose dexamethasone group, indicating a significant enhancement of sunitinib metabolism by dexamethasone. Moreover, the total levels of sunitinib and its metabolite are also significantly increased. The impact of interactions on sunitinib metabolism, as observed with CYP3A inducers such as dexamethasone, is a crucial consideration for clinical practice. To optimize the dosage and prevent adverse drug events, therapeutic drug monitoring can be employed to avoid the toxicity from such interactions.
Collapse
Affiliation(s)
- Guang-Rong Lu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rui-Zhen Wang
- Department of Pharmacy, Wenzhou People's Hospital, Wenzhou, Zhejiang, China
| | - Xin-Yu Zhao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jun-Er Xu
- Alberta Institute, Wenzhou Medical University, Zhejiang, China
| | - Cheng-Ke Huang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Sun
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rui-Jie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhe Wang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
7
|
Del Re M, Crucitta S, Brighi N, Kinspergher S, Mercinelli C, Rizzo M, Conteduca V, Rebuzzi SE, Beninato T, Venturi G, Doni L, Verzoni E, Puglisi S, Landriscina M, Porta C, Manfredi F, Caffo O, De Giorgi U, Fogli S, Danesi R. Concomitant Administration of VEGFR Tyrosine Kinase and Proton Pump Inhibitors May Impair Clinical Outcome of Patients With Metastatic Renal Cancer. Clin Genitourin Cancer 2024; 22:102147. [PMID: 39030142 DOI: 10.1016/j.clgc.2024.102147] [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: 12/08/2023] [Accepted: 06/23/2024] [Indexed: 07/21/2024]
Abstract
INTRODUCTION The administration of proton pump inhibitors (PPIs) is a common practice to reduce gastro-esophageal adverse events associated with drug treatments but may impair absorption and exposure to oncology drugs. This study investigated the effect of concomitant administration of PPIs and pazopanib, sunitinib and cabozantinib on survival of patients with metastatic clear cell renal carcinoma (mRCC). PATIENTS AND METHODS Total 451 patients receiving pazopanib, sunitinib and cabozantinib as first line treatment were enrolled in this retrospective study. Patients were defined as "no concomitant PPIs (PPI-)" if no PPIs were administered during TKIs, and as "concomitant PPIs (PPI+)" if the administration of PPIs was at least 75% of the time during which TKIs were given. RESULTS Eighty patients administered pazopanib were PPI- and 86 PPI+; no difference in PFS was observed (10.7 vs. 11.9 months, P = .79). If patients were stratified as short (n = 89) and long (n = 77) responders, there was a significant difference in terms of PFS in PPI+ (n = 47) versus PPI- (n = 30) in long responders, being 24.7 versus 38 months (P = .04), respectively. In the sunitinib cohort, no significant difference of PFS in PPI+ (n = 102) versus PPI- (n = 131) was found, being 11.3 versus 18.1 months, respectively (P=0.15). In the cabozantinib cohort, there was a statistically significant difference in PFS of PPI+ versus PPI- (6 months vs. not reached, P = .04). No correlation with adverse events was found. CONCLUSIONS This study demonstrates an association between PPIs and impaired PFS in mRCC patients given pazopanib and cabozantinib and recommends caution on their concomitant use.
Collapse
Affiliation(s)
- Marzia Del Re
- Department of Clinical and Experimental Medicine, Unit of Clinical Pharmacology and Pharmacogenetics, University of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Department of Clinical and Experimental Medicine, Unit of Clinical Pharmacology and Pharmacogenetics, University of Pisa, Pisa, Italy
| | - Nicole Brighi
- Oncology Department, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | | | - Chiara Mercinelli
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Mimma Rizzo
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Policlinico di Bari, Bari, Italy
| | - Vincenza Conteduca
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Sara Elena Rebuzzi
- Unit of Medical Oncology 1, Department of Internal Medicine and Medical Specialties, University of Genova, IRCCS Ospedale San Martino, Genova, Italy
| | - Teresa Beninato
- Department of Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Giulia Venturi
- Department of Oncology, Azienda Ospedaliero-Universitaria Careggi, Firenze, Italy
| | - Laura Doni
- Department of Oncology, Azienda Ospedaliero-Universitaria Careggi, Firenze, Italy
| | - Elena Verzoni
- Department of Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Silvia Puglisi
- Unit of Medical Oncology 1, Department of Internal Medicine and Medical Specialties, University of Genova, IRCCS Ospedale San Martino, Genova, Italy
| | - Matteo Landriscina
- Unit of Medical Oncology and Biomolecular Therapy, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Camillo Porta
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Policlinico di Bari, Bari, Italy; Interdisciplinary Department of Medicine, University of Bari "A. Moro", Bari, Italy
| | - Fiorella Manfredi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Orazio Caffo
- Department of Medical Oncology, Santa Chiara Hospital, Trento, Italy
| | - Ugo De Giorgi
- Oncology Department, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Stefano Fogli
- Department of Clinical and Experimental Medicine, Unit of Clinical Pharmacology and Pharmacogenetics, University of Pisa, Pisa, Italy
| | - Romano Danesi
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy.
| |
Collapse
|
8
|
Hu J, Xia H, Chen X, Xu X, Wu HL, Shen Y, Xu RA, Wu W. Effect of isavuconazole on the pharmacokinetics of sunitinib and its mechanism. BMC Cancer 2024; 24:1131. [PMID: 39261851 PMCID: PMC11389264 DOI: 10.1186/s12885-024-12904-4] [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: 04/09/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Sunitinib, a newly developed multi-targeted tyrosine kinase inhibitor (TKI), has become a common therapeutic option for managing advanced renal cell carcinoma (RCC). Examining the mechanism underlying the interaction between sunitinib and isavuconazole was the aim of this effort. METHODS The concentrations of sunitinib and its primary metabolite, N-desethyl sunitinib, were analyzed and quantified using ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). Our study evaluated the potential interaction between isavuconazole and sunitinib using rat liver microsomes (RLM), human liver microsomes (HLM), and in vivo rat models. For the in vivo study, two groups (n = 5) of Sprague-Dawley (SD) rats were randomly allocated to receive sunitinib either with or without co-administration of isavuconazole. Additionally, the effects of isavuconazole on the metabolic stability of sunitinib and N-desethyl sunitinib were studied in RLM in vitro. RESULTS Our findings demonstrated that in RLM, isavuconazole exhibited a mixed non-competitive and competitive inhibition mechanism, with an IC50 (half maximal inhibitory concentration) value of 1.33 µM. Meanwhile, in HLM, isavuconazole demonstrated a competitive inhibition mechanism, with an IC50 of 5.30 µM. In vivo studies showed that the presence of isavuconazole significantly increased the pharmacokinetic characteristics of sunitinib, with the AUC(0→t), AUC(0→∞), and Tmax rising to approximately 211.38%, 203.92%, and 288.89%, respectively, in contrast to the control group (5 mg/kg sunitinib alone). The pharmacokinetic characteristics of the metabolite N-desethyl sunitinib in the presence of isavuconazole remained largely unchanged compared to the control group. Furthermore, in vitro metabolic stability experiments revealed that isavuconazole inhibited the metabolic processing of both sunitinib and N-desethyl sunitinib. CONCLUSIONS Isavuconazole had a major impact on sunitinib metabolism, providing fundamental information for the precise therapeutic administration of sunitinib.
Collapse
Affiliation(s)
- Jinyu Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hailun Xia
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaohai Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinhao Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hua-Lu Wu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxin Shen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ren-Ai Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Wenzhi Wu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
9
|
Tisack A, Mohammad TF. Drug-Induced Pigmentation: A Review. Drugs 2024; 84:1071-1091. [PMID: 39085684 DOI: 10.1007/s40265-024-02062-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 08/02/2024]
Abstract
Drug-induced pigmentation (DIP) is estimated to account for 20% of all cases of acquired hyperpigmentation. Over 50 agents have been implicated, including antibiotics, antimalarials, antiretrovirals, antipsychotics, prostaglandin analogs, heavy metals, and chemotherapeutic agents. The skin, mucosal surfaces, nails, and hair can all be affected, with the color, distribution, onset, and duration of pigmentation varying between offending agents. Both a thorough physical examination and medication history are necessary to determine the offending agent. In terms of mechanism, DIP occurs most frequently through the accumulation of melanin within the dermis but also by drug accumulation, pigment synthesis, and iron deposition. Photoprotection, including applying a broad-spectrum sunscreen, wearing photoprotective clothing, and seeking shade, plays an important role in the prevention of exacerbation of DIP. Multiple lasers, including the picosecond alexandrite, Q-switched Nd:YAG, Q-switched alexandrite, and Q-switched ruby lasers, have been successful in obtaining clearance of DIP. In this review, we examine the unique characteristics of each of the inciting agents in terms of incidence, clinical presentation, time to onset and resolution, and pathogenesis.
Collapse
Affiliation(s)
- Aaron Tisack
- Department of Dermatology, Henry Ford Health, 3031 W Grand Blvd, Detroit, MI, 48202, USA
| | - Tasneem F Mohammad
- Department of Dermatology, Henry Ford Health, 3031 W Grand Blvd, Detroit, MI, 48202, USA.
| |
Collapse
|
10
|
Maruyama S, Momo K, Anno T, Ishida M, Kanno H, Kato M. The significance of therapeutic drug monitoring in detecting low medication adherence in patients with cancer: A case study of cabozantinib. Clin Case Rep 2024; 12:e9462. [PMID: 39308660 PMCID: PMC11413627 DOI: 10.1002/ccr3.9462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/15/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Maintaining good medication adherence is important for providing desirable outcomes from medication therapy. We showed that therapeutic drug monitoring (TDM) contributed to the identification of low medication adherence to cabozantinib in a patient with cancer. We present an educational case to assist with understanding TDM in a patient with cancer.
Collapse
Affiliation(s)
- Shinichi Maruyama
- Department of Bioanalytical ChemistryShowa University Graduate School of PharmacyShinagawa‐kuJapan
- Department of PharmacySaiseikai Yokohamashi Tobu HospitalYokohama cityJapan
| | - Kenji Momo
- Department of Hospital Pharmaceutics, School of PharmacyShowa UniversityShinagawa‐kuJapan
| | - Tadatsugu Anno
- Department of UrologySaiseikai Yokohamashi Tobu HospitalYokohama cityJapan
| | - Masaru Ishida
- Department of UrologySaiseikai Yokohamashi Tobu HospitalYokohama cityJapan
| | - Hiroshi Kanno
- Department of PharmacySaiseikai Yokohamashi Tobu HospitalYokohama cityJapan
| | - Masaru Kato
- Department of Bioanalytical ChemistryShowa University Graduate School of PharmacyShinagawa‐kuJapan
| |
Collapse
|
11
|
Maruyama S, Kobayashi H, Hiraga T, Anno T, Sanjo T, Arai M, Ishida M, Kanno H, Kato M. Association of Plasma Cabozantinib Concentration With Treatment Response and Adverse Events in Japanese Patients With Advanced Renal Cell Carcinoma. Ther Drug Monit 2024:00007691-990000000-00260. [PMID: 39208400 DOI: 10.1097/ftd.0000000000001254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/25/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Cabozantinib is highly effective against advanced renal cell carcinoma (RCC). However, approximately 60% of the patients require a dose reduction due to severe adverse events. Although associations between trough concentrations of cabozantinib and its efficacy and safety have been reported in other countries, reports on Japanese patients are unavailable. Therefore, we investigated the association of cabozantinib trough concentration with therapeutic efficacy and adverse events in Japanese patients with RCC and evaluated the usefulness of therapeutic drug monitoring. METHODS In this prospective observational study, we measured the trough concentrations of cabozantinib in 10 Japanese patients with RCC enrolled between May 2022 and September 2023. The associations of trough concentration with treatment response, as determined by RECIST 1.1, and the occurrence of grade 2 or higher adverse events were assessed. RESULTS Trough concentration was higher in patients with controlled cancer than in those with progressive cancer (1024 ± 352 versus 457 ± 216 ng/mL, P = 0.035). In addition, patients with grade 2 or higher adverse events showed a significantly higher trough concentration than those without (1560 ± 513 versus 807 ± 319 ng/mL, P = 0.032). In particular, grade 2 or higher dysgeusia, anorexia, fatigue, and dyspepsia significantly correlated with trough concentrations. CONCLUSIONS This is the first clinical study to demonstrate a correlation between cabozantinib trough concentration, therapeutic efficacy, and adverse events in Japanese patients with RCC. The therapeutic drug monitoring of cabozantinib could be useful for improving therapeutic efficacy and avoiding serious adverse events.
Collapse
Affiliation(s)
- Shinichi Maruyama
- Department of Bioanalytical Chemistry, Showa University Graduate School of Pharmacy, Tokyo, Japan
- Departments of Pharmacy and
| | - Hiroaki Kobayashi
- Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
- Department of Urology, National Defense Medical College Hospital, Saitama, Japan
| | | | - Tadatsugu Anno
- Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
| | - Tansei Sanjo
- Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
| | - Masashi Arai
- Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
| | - Masaru Ishida
- Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
| | | | - Masaru Kato
- Department of Bioanalytical Chemistry, Showa University Graduate School of Pharmacy, Tokyo, Japan
| |
Collapse
|
12
|
Sato Y, Kondo H, Sato Y, Abe A, Kikuchi M, Sato T, Kumondai M, Yoshikawa K, Hayakawa Y, Maekawa M, Mano N. Development of Simultaneous Drug Concentration Measurement Method Using an Automated Pretreatment Liquid Chromatography/Tandem Mass Spectrometry System for Therapeutic Drug Monitoring. Pharmaceutics 2024; 16:1138. [PMID: 39339175 PMCID: PMC11435224 DOI: 10.3390/pharmaceutics16091138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Therapeutic drug monitoring (TDM) is a personalized treatment approach that involves optimizing drug dosages based on patient-specific factors, such as drug plasma concentrations, therapeutic efficacy, or adverse reactions. The plasma concentration of drugs is determined using liquid chromatography/tandem mass spectrometry (LC-MS/MS) or various immunoassays. Compared with immunoassays, LC-MS/MS requires more pretreatment time as the number of samples increases. Recently, fully automated pretreatment LC-MS/MS systems have been developed to automatically perform whole-sample pretreatment for LC-MS/MS analysis. In this study, we developed a method for simultaneous concentration determination of five analytes (clozapine, mycophenolic acid, sunitinib, N-desethylsunitinib, and voriconazole) using LC-MS/MS for clinical TDM using a fully automated LC-MS/MS pretreatment system. In the developed method, the intra- and inter-assay relative error (RE) values ranged between -14.8% and 11.3%; the intra- and inter-assay coefficient of variation (CV) values were <8.8% and <10.5%, respectively. The analytes showed good stability, with RE values ranging between -13.6% and 10.9% and CV values <8.9%. Furthermore, the plasma concentrations in clinical samples using this method and the conventional manual pretreatment method showed similar results. Therefore, the method developed in this study could be considered a useful pretreatment method for routine TDM in patients.
Collapse
Affiliation(s)
- Yu Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Hiroki Kondo
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yuji Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Ai Abe
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Masafumi Kikuchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Toshihiro Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Masaki Kumondai
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Kohei Yoshikawa
- Shimadzu Corporation, 1 Nishinokyo Kuwabara-cho, Nakagyo-ku, Kyoto 604-8511, Japan
| | - Yoshihiro Hayakawa
- Shimadzu Corporation, 1 Nishinokyo Kuwabara-cho, Nakagyo-ku, Kyoto 604-8511, Japan
| | - Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| |
Collapse
|
13
|
Yang F, Lee G, Fan Y. Navigating tumor angiogenesis: therapeutic perspectives and myeloid cell regulation mechanism. Angiogenesis 2024; 27:333-349. [PMID: 38580870 PMCID: PMC11303583 DOI: 10.1007/s10456-024-09913-z] [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: 12/31/2023] [Accepted: 03/04/2024] [Indexed: 04/07/2024]
Abstract
Sustained angiogenesis stands as a hallmark of cancer. The intricate vascular tumor microenvironment fuels cancer progression and metastasis, fosters therapy resistance, and facilitates immune evasion. Therapeutic strategies targeting tumor vasculature have emerged as transformative for cancer treatment, encompassing anti-angiogenesis, vessel normalization, and endothelial reprogramming. Growing evidence suggests the dynamic regulation of tumor angiogenesis by infiltrating myeloid cells, such as macrophages, myeloid-derived suppressor cells (MDSCs), and neutrophils. Understanding these regulatory mechanisms is pivotal in paving the way for successful vasculature-targeted cancer treatments. Therapeutic interventions aimed to disrupt myeloid cell-mediated tumor angiogenesis may reshape tumor microenvironment and overcome tumor resistance to radio/chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Gloria Lee
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
14
|
Thakur A, Rana M, Mishra A, Kaur C, Pan CH, Nepali K. Recent advances and future directions on small molecule VEGFR inhibitors in oncological conditions. Eur J Med Chem 2024; 272:116472. [PMID: 38728867 DOI: 10.1016/j.ejmech.2024.116472] [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: 03/07/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024]
Abstract
"A journey of mixed emotions" is a quote that best describes the progress chart of vascular endothelial growth factor receptor (VEGFR) inhibitors as cancer therapeutics in the last decade. Exhilarated with the Food and Drug Administration (FDA) approvals of numerous VEGFR inhibitors coupled with the annoyance of encountering the complications associated with their use, drug discovery enthusiasts are on their toes with an unswerving determination to enhance the rate of translation of VEGFR inhibitors from preclinical to clinical stage. The recently crafted armory of VEGFR inhibitors is a testament to their growing dominance over other antiangiogenic therapies for cancer treatment. This review perspicuously underscores the earnest attempts of the researchers to extract the antiproliferative potential of VEGFR inhibitors through the design of mechanistically diverse structural assemblages. Moreover, this review encompasses sections on structural/molecular properties and physiological functions of VEGFR, FDA-approved VEGFR inhibitors, and hurdles restricting the activity range/clinical applicability of VEGFR targeting antitumor agents. In addition, tactics to overcome the limitations of VEGFR inhibitors are discussed. A clear-cut viewpoint transmitted through this compilation can provide practical directions to push the cart of VEGFR inhibitors to advanced-stage clinical investigations in diverse malignancies.
Collapse
Affiliation(s)
- Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Mandeep Rana
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Anshul Mishra
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Charanjit Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Chun-Hsu Pan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
15
|
Giraud EL, Westerdijk K, van der Kleij MBA, Guchelaar NAD, Meertens M, Bleckman RF, Rieborn A, Mohammadi M, Roets E, Mathijssen RHJ, Huitema ADR, Koolen SLW, Gelderblom H, Moes DJAR, Reyners AKL, Touw DJ, Keizer-Heldens P, Oosten AW, van der Graaf WTA, Steeghs N, van Erp NP, Desar IME. Sunitinib for the treatment of metastatic gastrointestinal stromal tumors: the effect of TDM-guided dose optimization on clinical outcomes. ESMO Open 2024; 9:103477. [PMID: 38833964 PMCID: PMC11179075 DOI: 10.1016/j.esmoop.2024.103477] [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: 02/01/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Sunitinib is an oral anticancer drug approved for the treatment of among others gastrointestinal stromal tumor (GIST). Previous analyses demonstrated an exposure-response relationship at the standard dose, and minimum target levels of drug exposure have been defined above which better treatment outcomes are observed. Therapeutic drug monitoring (TDM) could be used as a tool to optimize the individual dose, aiming at sunitinib trough concentrations ≥37.5 ng/ml for continuous dosing. Nonetheless, data on the added value of TDM-guided dosing on clinical endpoints are currently lacking. Therefore, we evaluate the effect of TDM in patients with advanced and metastatic GIST treated with sunitinib in terms of efficacy and toxicity. PATIENTS AND METHODS A TDM-guided cohort was compared to a non-TDM-guided cohort in terms of median progression-free survival (mPFS) and overall survival (mOS). Also, mPFS between patients with and without dose-limiting toxicities (DLTs) was compared. Patients in the prospective cohort were included in two studies on TDM-guided dosing (the DPOG-TDM study and TUNE study). The retrospective cohort consisted of patients from the Dutch GIST Registry who did not receive TDM-guided dosing. RESULTS In total, 51 and 106 patients were included in the TDM-guided cohort and non-TDM-guided cohort, respectively. No statistical difference in mPFS was observed between these two cohorts (39.4 versus 46.9 weeks, respectively; P = 0.52). Patients who experienced sunitinib-induced DLTs had longer mPFS compared to those who did not (51.9 versus 28.9 weeks, respectively; P = 0.002). CONCLUSIONS Our results do not support the routine use of TDM-guided dose optimization of sunitinib in patients with advanced/metastatic GIST to improve survival.
Collapse
Affiliation(s)
- E L Giraud
- Department of Pharmacy, Radboud University Medical Center, Radboud University Medical Center, Nijmegen, The Netherlands. https://twitter.com/ElineGirau45556
| | - K Westerdijk
- Department of Medical Oncology, Radboud University Medical Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M B A van der Kleij
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - N A D Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - M Meertens
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R F Bleckman
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A Rieborn
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - M Mohammadi
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - E Roets
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - A D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - S L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands; Department of Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - H Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - D J A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - A K L Reyners
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - D J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - P Keizer-Heldens
- Department of Medical Oncology, Rijnstate Hospital, Arnhem, The Netherlands
| | - A W Oosten
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - W T A van der Graaf
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - N Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - N P van Erp
- Department of Pharmacy, Radboud University Medical Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I M E Desar
- Department of Medical Oncology, Radboud University Medical Center, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
16
|
Zhao Y, Zhang X, Ding X, Wang Y, Li Z, Zhao R, Cheng HE, Sun Y. Efficacy and safety of FLT3 inhibitors in monotherapy of hematological and solid malignancies: a systemic analysis of clinical trials. Front Pharmacol 2024; 15:1294668. [PMID: 38828446 PMCID: PMC11140126 DOI: 10.3389/fphar.2024.1294668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/29/2024] [Indexed: 06/05/2024] Open
Abstract
Introduction: FLT3 mutations are closely associated with the occurrence of hematological and solid malignancies, especially with acute myeloid leukemia. Currently, several FLT3 inhibitors are in clinical trials, and some have been applied in clinic. However, the safety, efficacy and pharmacodynamics of these FLT3 inhibitors have not been systemically analyzed before. Methods: We searched and reviewed clinical trial reports on the monotherapy of 13 FLT3 inhibitors, including sorafenib, lestaurtinib, midostaurin, gilteritinib, quizartinib, sunitinib, crenolanib, tandutinib, cabozantinib, pexidartinib, pacritinib, famitinib, and TAK-659 in patients with hematological and solid malignancies before May 31, 2023. Results: Our results showed the most common adverse events (AEs) were gastrointestinal adverse reactions, including diarrhea, hand-foot syndrome and nausea, while the most common hematological AEs were febrile neutropenia, anemia, and thrombocytopenia. Based on the published data, the mean overall survival (OS) and the mean progression-free survival (PFS) were 9.639 and 5.905 months, respectively. The incidence of overall response rate (ORR), complete remission (CR), partial response (PR), and stable disease (SD) for all these FLT3 inhibitors was 29.0%, 8.7%, 16.0%, and 42.3%, respectively. The ORRs of FLT3 inhibitors in hematologic malignancies and solid tumors were 40.8% and 18.8%, respectively, indicating FLT3 inhibitors were more effective for hematologic malignancies than for solid tumors. In addition, time to maximum plasma concentration (Tmax) in these FLT3 inhibitors ranged from 0.7-12.0 hours, but the elimination half-life (T1/2) range was highly variable, from 6.8 to 151.8 h. Discussion: FLT3 inhibitors monotherapy has shown significant anti-tumor effect in clinic, and the effectiveness may be further improved through combination medication.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hai-En Cheng
- School of Medical Laboratory, Shandong Second Medical University, Weifang, China
| | - Yanli Sun
- School of Medical Laboratory, Shandong Second Medical University, Weifang, China
| |
Collapse
|
17
|
Lin L, van der Meer EKO, Steeghs N, Beijnen JH, Huitema ADR. Are novel oral oncolytics underdosed in obese patients? Cancer Chemother Pharmacol 2024; 93:129-136. [PMID: 37906253 PMCID: PMC10853358 DOI: 10.1007/s00280-023-04601-z] [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: 08/14/2023] [Accepted: 10/05/2023] [Indexed: 11/02/2023]
Abstract
PURPOSE Data on the effects of obesity on drug exposure of oral targeted oncolytics is scarce. Therefore, the aim of this study was to investigate the influence of body weight and body mass index (BMI) on trough levels of oral oncolytics with an exposure-response relationship. The oral oncolytics of interest were abiraterone, alectinib, cabozantinib, crizotinib, imatinib, pazopanib, sunitinib and trametinib. METHODS This retrospective cohort study included patients treated with the selected oral oncolytics at the standard dose, with a measured trough level at steady state and with available body weight. The Spearman's correlation test was used to determine the correlation between body weight and trough levels. The Fisher's exact text was used to compare the frequency of inadequate trough levels between BMI categories. RESULTS 1265 patients were included across the different oral oncolytics. A negative correlation coefficient was observed between weight and trough levels for crizotinib (n = 75), imatinib (n = 201) and trametinib (n = 310), respectively, ρ = - 0.41, ρ = - 0.24 and ρ = - 0.23, all with a p-value < 0.001. For crizotinib, a higher percentage of patients with a body weight > 100 kg had inadequate trough levels. No statistically significant differences were observed in the frequency of inadequate trough levels between BMI categories. CONCLUSION Higher body weight was only correlated with lower plasma trough levels for crizotinib, imatinib, and trametinib. Therefore, patients with a high body weight may require dose escalation to obtain adequate target levels when treated with these oral oncolytics.
Collapse
Affiliation(s)
- Lishi Lin
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Ellen K O van der Meer
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
18
|
Kumari R, Syeda S, Shrivastava A. Nature's Elixir for Cancer Treatment: Targeting Tumor-induced Neovascularization. Curr Med Chem 2024; 31:5281-5304. [PMID: 38425113 DOI: 10.2174/0109298673282525240222050051] [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: 10/17/2023] [Revised: 01/20/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Angiogenesis, a multistep process, involves sprouting of new vessels from the pre-existing vessels in response to a stimulus in its microenvironment. Normally, angiogenesis is important for tissue maintenance and homeostasis, however it is also known to be associated with various pathologies, including cancer. Importantly, neovascularization is very crucial for tumors to grow and metastasize since it allows delivery of oxygen and nutrients as well as promotes tumor cell dissemination to distant sites. Activation of angiogenic switch is a consequence of imbalance in pro- as well as anti-angiogenic factors, that are immensely impacted by reactive oxygen species and epigenetic regulation. Several reports have suggested that angiogenic inhibitors significantly inhibit tumor growth. Therefore, anti-angiogenic therapy has gained substantial attention and has been considered a rational approach in cancer therapeutics. In this line, several anti- angiogenic drugs have been approved, however, their long term usage caused several side effects. In view of this, researchers switched to plant-based natural compounds for identifying safe and cost-effective anti-angiogenic drugs. Of note, various phytochemicals have been evaluated to reduce tumor growth by inhibiting tumor-induced angiogenesis. Moreover, the implication of nano-carriers to enhance the bioavailability of phytochemicals has proven to be more efficient anti-cancer agents. The present review highlights the existing knowledge on tumor-induced neovascularization and its regulation at the epigenetic level. Further, we emphasize the inhibitory effect of phytochemicals on tumor- induced angiogenesis that will open up new avenues in cancer therapeutics.
Collapse
Affiliation(s)
- Rani Kumari
- Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Saima Syeda
- Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Anju Shrivastava
- Department of Zoology, University of Delhi, Delhi, 110007, India
| |
Collapse
|
19
|
Li Q, Tang T, Zhang M, Li L, Chen W. An Optimized LC-MS/MS Method for Quantification of Sunitinib and N -Desethyl Sunitinib in Human Plasma and Its Application for Therapeutic Drug Monitoring. Ther Drug Monit 2023; 45:817-822. [PMID: 37074815 DOI: 10.1097/ftd.0000000000001097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/04/2023] [Indexed: 04/20/2023]
Abstract
BACKGROUND Sunitinib (SUN) malate is an oral, multitargeted, tyrosine kinase inhibitor approved for the treatment of metastatic renal cell carcinoma, imatinib-resistant or imatinib-intolerant gastrointestinal stromal tumors, and pancreatic neuroendocrine tumors. SUN has a narrow therapeutic window and high variability in interpatient pharmacokinetic parameters. Clinical detection methods for SUN and N -desethyl SUN limit the application of SUN to therapeutic drug monitoring. All published methods for quantifying SUN in human plasma require strict light protection to avoid light-induced isomerism or the use of additional quantitative software. To avoid these difficult processes in clinical routines, the authors propose a novel method that merges the peaks of the E -isomer and Z -isomer of SUN or N -desethyl SUN into a single peak. METHODS The E -isomer and Z -isomer peaks of SUN or N -desethyl SUN were merged into a single peak by optimizing the mobile phases to decrease the resolution of the isomers. A suitable chromatographic column was selected to obtain a good peak shape. Thereafter, the conventional and single-peak methods (SPM) were simultaneously validated and compared according to the guidelines published by the Food and Drug Administration in 2018 and the Chinese Pharmacopoeia in 2020. RESULTS The verification results showed that the SPM was superior to the conventional method in the matrix effect and met the requirements for biological sample analysis. SPM was then applied to detect the total steady-state concentration of SUN and N -desethyl SUN in tumor patients who received SUN malate. CONCLUSIONS The established SPM makes the detection of SUN and N -desethyl SUN easier and faster without light protection or extra quantitative software, making it more appropriate for routine clinical use. The clinical application results showed that 12 patients took 37.5 mg per day, with a median total trough steady-state concentration of 75.0 ng/mL.
Collapse
Affiliation(s)
- Qiaoqiao Li
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing, China
| | | | | | | | | |
Collapse
|
20
|
El Sharkasy ME, Tolba MM, Belal F, Walash MI, AboShabana R. Utility of the food colorant erythrosine B as an effective green probe for quantitation of the anticancer sunitinib. Application to pharmaceutical formulations and human plasma. LUMINESCENCE 2023; 38:2073-2085. [PMID: 37747151 DOI: 10.1002/bio.4598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/30/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023]
Abstract
Sunitinib is a tyrosine kinase inhibitor used for the treatment of renal cell carcinoma and gastrointestinal stromal tumors. In this study, two spectroscopic methods, spectrofluorometric and spectrophotometric, were utilized to quantify sunitinib in different matrices. In method I, the native fluorescence of erythrosine B was quenched by forming ion-pair complex with increasing quantities of sunitinib. This approach was utilized for measuring sunitinib in its dosage forms and spiked plasma. After excitation at 528 nm, the quenching of fluorescence is linearly related to the concentration across the range of 0.05-0.5 μg mL-1 at 550 nm in Britton-Robinson buffer (pH 4.0), with a correlation value of 0.9999 and a high level of sensitivity with detection limit down to 10 ng mL-1 . Method II relies on spectrophotometric measurements of the produced complex at 550 nm across a range of 0.5-10.0 μg mL-1 , with good correlation value of 0.9999. This method has a detection limit down to 0.16 μg mL-1 . The proposed methodologies were validated according to International Conference on Harmonization (ICH) guidelines with satisfactory results. The stoichiometry of the reaction was determined through the application of Job's method, while the mechanism of quenching was investigated by employing the Stern-Volmer plot. The designated methods were used to estimate sunitinib in its capsules and in spiked human plasma. Additionally, the statistical analysis of the data revealed no substantial differences when compared to previous reported spectroscopic method. Green assessment tools provide further details about the eco-friendly nature of the methods.
Collapse
Affiliation(s)
- Mona E El Sharkasy
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Manar M Tolba
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Fathalla Belal
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed I Walash
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Rasha AboShabana
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
21
|
Jiang H, Wang X, Guo L, Tan X, Gui X, Liao Z, Li Z, Chen X, Wu X. Effect of sunitinib against Echinococcus multilocularis through inhibition of VEGFA-induced angiogenesis. Parasit Vectors 2023; 16:407. [PMID: 37936208 PMCID: PMC10631006 DOI: 10.1186/s13071-023-05999-4] [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: 03/07/2023] [Accepted: 10/04/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Alveolar echinococcosis (AE) is a lethal zoonosis caused by the fox tapeworm Echinococcus multilocularis. The disease is difficult to treat, and an effective therapeutic drug is urgently needed. Echinococcus multilocularis-associated angiogenesis is required by the parasite for growth and metastasis; however, whether antiangiogenic therapy is effective for treating AE is unclear. METHODS The in vivo efficacy of sunitinib malate (SU11248) was evaluated in mice by secondary infection with E. multilocularis. Enzyme-linked immunosorbent assays (ELISAs) were used to evaluate treatment effects on serum IL-4 and vascular endothelial growth factor A (VEGFA) levels after SU11248 treatment. Gross morphological observations and immunohistochemical staining were used to evaluate the impact of SU11248 on angiogenesis and the expression of pro-angiogenic factors VEGFA and VEGF receptor 2 (VEGFR2) in the metacestode tissues. Furthermore, the anthelmintic effects of SU11248 were tested on E. multilocularis metacestodes in vitro. The effect of SU11248 on the expression of VEGFA, VEGFR2, and phosphorylated VEGFR2 (p-VEGFR2) in liver cells infected with protoscoleces in vitro was detected by western blotting, reverse transcription quantitative polymerase chain reaction (RT-qPCR), and enzyme-linked immunosorbent assay (ELISA). The influence of SU11248 on endothelial progenitor cell (EPC) proliferation and migration was determined using CCK8 and transwell assays. RESULTS In vivo, SU11248 treatment markedly reduced neovascular lesion formation and substantially inhibited E. multilocularis metacestode growth in mice. Further, it exhibited high anti-hydatid activity as efficiently as albendazole (ABZ), and the treatment resulted in reduced protoscolex development. In addition, VEGFA, VEGFR2, and p-VEGFR2 expression was significantly decreased in the metacestode tissues after SU11248 treatment. However, no effect of SU11248 on serum IL-4 levels was observed. In vitro, SU11248 exhibited some anthelmintic effects and damaged the cellular structure in the germinal layer of metacestodes at concentrations below those generally considered acceptable for treatment (0.12-0.5 μM). Western blotting, RT-qPCR, and ELISA showed that in co-cultured systems, only p-VEGFR2 levels tended to decrease with increasing SU11248 concentrations. Furthermore, SU11248 was less toxic to Reuber rat hepatoma (RH) cells and metacestodes than to EPCs, and 0.1 μM SU11248 completely inhibited EPC migration to the supernatants of liver cell and protoscolex co-cultures. CONCLUSIONS SU11248 is a potential candidate drug for the treatment of AE, which predominantly inhibits parasite-induced angiogenesis. Host-targeted anti-angiogenesis treatment strategies constitute a new avenue for the treatment of AE.
Collapse
Affiliation(s)
- Huijiao Jiang
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Xiaoyi Wang
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Lijiao Guo
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Xiaowu Tan
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Xianwei Gui
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Zhenyu Liao
- Department of Immunology, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
- Department of Experimental Medicine, Jintang First People's Hospital West China Hospital Sichuan University Jintang Hospital, Chengdu, 610400, Sichuan, China
| | - Zhiwei Li
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Xueling Chen
- Department of Immunology, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China.
| | - Xiangwei Wu
- National Health Commission of the People's Republic of China Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China.
| |
Collapse
|
22
|
Hayakawa T, Funakoshi S, Hamamoto Y, Hirata K, Kanai T. Sunitinib-induced hyperammonemic encephalopathy in metastatic gastrointestinal stromal tumors: A case report. World J Clin Cases 2023; 11:7629-7634. [DOI: 10.12998/wjcc.v11.i31.7629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/10/2023] [Accepted: 10/23/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Sunitinib, a multi-targeted tyrosine kinase inhibitor (TKI), has been approved for the salvage treatment of gastrointestinal stromal tumors (GIST). Hyperammonemic encephalopathy is a rare but severe complication of sunitinib use. Here, we present the case of a 66-year-old male with metastatic GIST without underlying liver cirrhosis who developed sunitinib-induced hyperammonemic encephalopathy.
CASE SUMMARY A 66-year-old male with metastatic GIST was admitted because of reduced consciousness. Imatinib was administered as the first-line systemic therapy. He experienced repeated episodes of peritonitis due to tumor perforation, and surgery was performed. Progressive disease was confirmed based on increased liver metastasis, and sunitinib was initiated as a salvage treatment. However, 23 d after the third course of sunitinib, he presented to the emergency room with an episode of altered consciousness and behavioral changes. Based on the patient clinical history and examination findings, sunitinib-induced encephalopathy was suspected. Sunitinib was discontinued, and the patient was treated for hyperammonemia. The patient had a normal level of consciousness four days later, and the serum ammonia level gradually decreased. No further neurological symptoms were reported in subsequent follow-ups.
CONCLUSION TKI-induced hyperammonemic encephalopathy is potentially life-threatening. Patients receiving TKIs experiencing adverse reactions should undergo systemic evaluation and prompt treatment.
Collapse
Affiliation(s)
- Takaoki Hayakawa
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Shinjuku-ku 160-8582, Tokyo, Japan
| | - Shinsuke Funakoshi
- Division of Medical Oncology, Department of Internal Medicine, Tokyo Saiseikai Central Hospital, Minato-ku 108-0073, Tokyo, Japan
| | - Yasuo Hamamoto
- Keio Cancer Center, Keio University School of Medicine, Shinjuku-ku 160-8582, Tokyo, Japan
| | - Kenro Hirata
- Keio Cancer Center, Keio University School of Medicine, Shinjuku-ku 160-8582, Tokyo, Japan
| | - Takanori Kanai
- Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku 160-8582, Tokyo, Japan
| |
Collapse
|
23
|
Liu C, Leighow SM, McIlroy K, Lu M, Dennis KA, Abello K, Brown DJ, Moore CJ, Shah A, Inam H, Rivera VM, Pritchard JR. Excessive concentrations of kinase inhibitors in translational studies impede effective drug repurposing. Cell Rep Med 2023; 4:101227. [PMID: 37852183 PMCID: PMC10591048 DOI: 10.1016/j.xcrm.2023.101227] [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: 03/07/2023] [Revised: 06/20/2023] [Accepted: 09/13/2023] [Indexed: 10/20/2023]
Abstract
Drug repositioning seeks to leverage existing clinical knowledge to identify alternative clinical settings for approved drugs. However, repositioning efforts fail to demonstrate improved success rates in late-stage clinical trials. Focusing on 11 approved kinase inhibitors that have been evaluated in 139 repositioning hypotheses, we use data mining to characterize the state of clinical repurposing. Then, using a simple experimental correction with human serum proteins in in vitro pharmacodynamic assays, we develop a measurement of a drug's effective exposure. We show that this metric is remarkably predictive of clinical activity for a panel of five kinase inhibitors across 23 drug variant targets in leukemia. We then validate our model's performance in six other kinase inhibitors for two types of solid tumors: non-small cell lung cancer (NSCLC) and gastrointestinal stromal tumors (GISTs). Our approach presents a straightforward strategy to use existing clinical information and experimental systems to decrease the clinical failure rate in drug repurposing studies.
Collapse
Affiliation(s)
- Chuan Liu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Scott M Leighow
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Kyle McIlroy
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Mengrou Lu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kady A Dennis
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kerry Abello
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Donovan J Brown
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Connor J Moore
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Anushka Shah
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Haider Inam
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | | | - Justin R Pritchard
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; The Huck Institute for the Life Sciences, Center for Resistance Evolution, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
24
|
van der Kleij MBA, Guchelaar NAD, Mathijssen RHJ, Versluis J, Huitema ADR, Koolen SLW, Steeghs N. Therapeutic Drug Monitoring of Kinase Inhibitors in Oncology. Clin Pharmacokinet 2023; 62:1333-1364. [PMID: 37584840 PMCID: PMC10519871 DOI: 10.1007/s40262-023-01293-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 08/17/2023]
Abstract
Although kinase inhibitors (KI) frequently portray large interpatient variability, a 'one size fits all' regimen is still often used. In the meantime, relationships between exposure-response and exposure-toxicity have been established for several KIs, so this regimen could lead to unnecessary toxicity and suboptimal efficacy. Dose adjustments based on measured systemic pharmacokinetic levels-i.e., therapeutic drug monitoring (TDM)-could therefore improve treatment efficacy and reduce the incidence of toxicities. Therefore, the aim of this comprehensive review is to give an overview of the available evidence for TDM for the 77 FDA/EMA kinase inhibitors currently approved (as of July 1st, 2023) used in hematology and oncology. We elaborate on exposure-response and exposure-toxicity relationships for these kinase inhibitors and provide practical recommendations for TDM and discuss corresponding pharmacokinetic targets when possible.
Collapse
Affiliation(s)
- Maud B A van der Kleij
- Division of Medical Oncology, Department of Clinical Pharmacology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, The Netherlands.
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| | - Niels A D Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jurjen Versluis
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Pharmacy, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Neeltje Steeghs
- Division of Medical Oncology, Department of Clinical Pharmacology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Jackson KD, Achour B, Lee J, Geffert RM, Beers JL, Latham BD. Novel Approaches to Characterize Individual Drug Metabolism and Advance Precision Medicine. Drug Metab Dispos 2023; 51:1238-1253. [PMID: 37419681 PMCID: PMC10506699 DOI: 10.1124/dmd.122.001066] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 07/09/2023] Open
Abstract
Interindividual variability in drug metabolism can significantly affect drug concentrations in the body and subsequent drug response. Understanding an individual's drug metabolism capacity is important for predicting drug exposure and developing precision medicine strategies. The goal of precision medicine is to individualize drug treatment for patients to maximize efficacy and minimize drug toxicity. While advances in pharmacogenomics have improved our understanding of how genetic variations in drug-metabolizing enzymes (DMEs) affect drug response, nongenetic factors are also known to influence drug metabolism phenotypes. This minireview discusses approaches beyond pharmacogenetic testing to phenotype DMEs-particularly the cytochrome P450 enzymes-in clinical settings. Several phenotyping approaches have been proposed: traditional approaches include phenotyping with exogenous probe substrates and the use of endogenous biomarkers; newer approaches include evaluating circulating noncoding RNAs and liquid biopsy-derived markers relevant to DME expression and function. The goals of this minireview are to 1) provide a high-level overview of traditional and novel approaches to phenotype individual drug metabolism capacity, 2) describe how these approaches are being applied or can be applied to pharmacokinetic studies, and 3) discuss perspectives on future opportunities to advance precision medicine in diverse populations. SIGNIFICANCE STATEMENT: This minireview provides an overview of recent advances in approaches to characterize individual drug metabolism phenotypes in clinical settings. It highlights the integration of existing pharmacokinetic biomarkers with novel approaches; also discussed are current challenges and existing knowledge gaps. The article concludes with perspectives on the future deployment of a liquid biopsy-informed physiologically based pharmacokinetic strategy for patient characterization and precision dosing.
Collapse
Affiliation(s)
- Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Brahim Achour
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Jonghwa Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Raeanne M Geffert
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Jessica L Beers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Bethany D Latham
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (K.D.J., J.L., R.M.G., J.L.B., B.D.L.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island (B.A.)
| |
Collapse
|
26
|
Zhang Y, Popel AS, Bazzazi H. Combining Multikinase Tyrosine Kinase Inhibitors Targeting the Vascular Endothelial Growth Factor and Cluster of Differentiation 47 Signaling Pathways Is Predicted to Increase the Efficacy of Antiangiogenic Combination Therapies. ACS Pharmacol Transl Sci 2023; 6:710-726. [PMID: 37200806 PMCID: PMC10186363 DOI: 10.1021/acsptsci.3c00008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Indexed: 05/20/2023]
Abstract
Angiogenesis is a critical step in tumor growth, development, and invasion. Nascent tumor cells secrete vascular endothelial growth factor (VEGF) that significantly remodels the tumor microenvironment through interaction with multiple receptors on vascular endothelial cells, including type 2 VEGF receptor (VEGFR2). The complex pathways initiated by VEGF binding to VEGFR2 lead to enhanced proliferation, survival, and motility of vascular endothelial cells and formation of a new vascular network, enabling tumor growth. Antiangiogenic therapies that inhibit VEGF signaling pathways were among the first drugs that targeted stroma rather than tumor cells. Despite improvements in progression-free survival and higher response rates relative to chemotherapy in some types of solid tumors, the impact on overall survival (OS) has been limited, with the majority of tumors eventually relapsing due to resistance or activation of alternate angiogenic pathways. Here, we developed a molecularly detailed computational model of endothelial cell signaling and angiogenesis-driven tumor growth to investigate combination therapies targeting different nodes of the endothelial VEGF/VEGFR2 signaling pathway. Simulations predicted a strong threshold-like behavior in extracellular signal-regulated kinases 1/2 (ERK1/2) activation relative to phosphorylated VEGFR2 levels, as continuous inhibition of at least 95% of receptors was necessary to abrogate phosphorylated ERK1/2 (pERK1/2). Combinations with mitogen-activated protein kinase/ERK kinase (MEK) and spingosine-1-phosphate inhibitors were found to be effective in overcoming the ERK1/2 activation threshold and abolishing activation of the pathway. Modeling results also identified a mechanism of resistance whereby tumor cells could reduce pERK1/2 sensitivity to inhibitors of VEGFR2 by upregulation of Raf, MEK, and sphingosine kinase 1 (SphK1), thus highlighting the need for deeper investigation of the dynamics of the crosstalk between VEGFR2 and SphK1 pathways. Inhibition of VEGFR2 phosphorylation was found to be more effective at blocking protein kinase B, also known as AKT, activation; however, to effectively abolish AKT activation, simulations identified Axl autophosphorylation or the Src kinase domain as potent targets. Simulations also supported activating cluster of differentiation 47 (CD47) on endothelial cells as an effective combination partner with tyrosine kinase inhibitors to inhibit angiogenesis signaling and tumor growth. Virtual patient simulations supported the effectiveness of CD47 agonism in combination with inhibitors of VEGFR2 and SphK1 pathways. Overall, the rule-based system model developed here provides new insights, generates novel hypothesis, and makes predictions regarding combinations that may enhance the OS with currently approved antiangiogenic therapies.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Aleksander S. Popel
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Hojjat Bazzazi
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
27
|
Wheless M, Das S. Systemic Therapy for Pancreatic Neuroendocrine Tumors. Clin Colorectal Cancer 2023; 22:34-44. [PMID: 36114085 DOI: 10.1016/j.clcc.2022.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/21/2022] [Accepted: 08/02/2022] [Indexed: 11/03/2022]
Abstract
Patients with metastatic or advanced pancreatic neuroendocrine tumors (NETs) carry poorer prognoses relative to patients with other NETs due to bulkier and often, more proliferative baseline disease. Patients with these tumors also possess more approved treatment options relative to patients with other NETs, making therapeutic sequencing nuanced. As such, defining optimal therapeutic sequencing and developing more potent cytoreductive treatments for patients are significant areas of research need in the field. Herein this review, we discuss the current systemic therapy landscape, our approach to therapeutic sequencing in the clinic and ongoing studies seeking to define optimal sequencing of systemic therapies, and novel therapeutics in development, for patients with pancreatic NETs. We limit the scope of this latter topic to agents with preclinical or clinical rationale over the last 8 years to provide a contemporary view of the drug development landscape and focus primarily on new types of peptide receptor radionuclide therapy, anti-vascular endothelial growth factor receptor tyrosine kinase inhibitors and anti-vascular endothelial growth receptor tyrosine kinase inhibitor plus immunotherapy combinations.
Collapse
Affiliation(s)
- Margaret Wheless
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN
| | - Satya Das
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology and Oncology, Nashville, TN.
| |
Collapse
|
28
|
Maruyama S, Kato M, Hiraga T, Ishida M, Kanno H. Quantitative determination of plasma cabozantinib concentration using HPLC-UV and its application to patients with renal cell carcinoma. Biomed Chromatogr 2023; 37:e5599. [PMID: 36760165 DOI: 10.1002/bmc.5599] [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: 12/24/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
Cabozantinib is an oral small-molecule tyrosine kinase inhibitor that has become a standard of care for advanced renal cell carcinoma (RCC). However, cabozantinib is associated with a high rate of adverse events. Therefore, individualised cabozantinib administration and monitoring could help maximise its therapeutic efficacy and avoid serious adverse events. This study developed and validated a method to determine cabozantinib concentration in plasma using HPLC-UV. Sorafenib, an internal standard, was added to the plasma sample containing cabozantinib. A calibration curve for cabozantinib showed good linearity (R2 = 1.00), between 25 and 4,000 ng/ml. The recovery rate was above 92.1%, and the intra- and inter-day coefficients of variation were smaller than 5.2 and 6.8%, respectively. Then, we applied the method for monitoring cabozantinib blood levels in three patients with advanced RCC who were taking cabozantinib at a dose of 20, 40 or 60 mg/day. Grade 3 adverse events were more likely to occur in patients with high dosing and blood level of cabozantinib. Owing to its simplicity, the developed method can be used in general hospitals, and is expected to help maximise drug efficacy and minimise serious adverse events in many patients with RCC undergoing cabozantinib treatment.
Collapse
Affiliation(s)
- Shinichi Maruyama
- Department of Pharmacy, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Kanagawa, Japan.,Division of Bioanalytical Chemistry, Graduate School of Pharmacy, Tokyo, Japan
| | - Masaru Kato
- Division of Bioanalytical Chemistry, Graduate School of Pharmacy, Tokyo, Japan
| | - Tatsuru Hiraga
- Department of Pharmacy, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Kanagawa, Japan
| | - Masaru Ishida
- Department of Urology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Kanagawa, Japan
| | - Hiroshi Kanno
- Department of Pharmacy, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Kanagawa, Japan
| |
Collapse
|
29
|
Sasaki K, Kanda T, Matsumoto Y, Ishikawa T, Hirota S, Saijo Y. Sunitinib therapy for imatinib-resistant and/or intolerant gastrointestinal stromal tumors: comparison of safety and efficacy between standard and reduced dosage regimens. Jpn J Clin Oncol 2023; 53:297-303. [PMID: 36644881 DOI: 10.1093/jjco/hyac202] [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: 06/28/2022] [Accepted: 12/11/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Sunitinib therapy for patients with imatinib-resistant and/or intolerant gastrointestinal stromal tumors (GISTs) often causes severe adverse events (AEs) that lead to treatment discontinuation. METHODS We retrospectively reviewed the clinical records of imatinib-resistant and/or intolerant GIST patients who underwent sunitinib therapy in our institutions between 2007 and 2020. Forty-one patients were enrolled and divided into two groups on the basis of the starting dosage: the standard dosage group (50 mg/day, 21 patients) and the reduced dosage group (37.5 mg/day, 20 patients). Tolerability, safety and clinical efficacy of the two groups were compared. RESULTS Three patients (14%) in the standard dosage group and another three (15%) in the reduced dosage group (P = 1.000) discontinued sunitinib therapy because of AEs. The incidences of grade 3 or more severe treatment-related AEs were 90 and 75%, respectively (P = 0.238). Two possible treatment-related deaths were noted in the standard dosage group. Clinical efficacy was comparable between the two groups: median time to treatment failure and overall survival were 4.5 months [interquartile range (IQR), 3.6-9.0] and 13.7 months (IQR, 7.5-22.9) in the standard dosage group and 4.6 months (IQR, 2.7-17.0) and 13.4 months (IQR, 9.3-36.8) in the reduced dosage group, respectively. CONCLUSIONS The reduced dosage of 37.5 mg sunitinib tended to decrease toxicity and the incidences of severe AEs and treatment-related deaths. This reduced dosage regimen showed equivalent clinical efficacy including patient survival. The reduced dosage of 37.5 mg sunitinib can be adopted as an alternative therapy for patients with imatinib-resistant and/or intolerant GISTs.
Collapse
Affiliation(s)
- Kenta Sasaki
- Department of Medical Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Department of Internal Medicine, Sanjo General Hospital, Sanjo, Japan
| | - Tatsuo Kanda
- Department of Surgery, Sanjo General Hospital, Sanjo, Japan
| | - Yoshifumi Matsumoto
- Department of Medical Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takashi Ishikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Japan
| | - Yasuo Saijo
- Department of Medical Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
30
|
Puisset F, Mseddi M, Mourey L, Pouessel D, Blanchet B, Chatelut E, Chevreau C. Therapeutic Drug Monitoring of Tyrosine Kinase Inhibitors in the Treatment of Advanced Renal Cancer. Cancers (Basel) 2023; 15:cancers15010313. [PMID: 36612311 PMCID: PMC9818258 DOI: 10.3390/cancers15010313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
Seven tyrosine kinase inhibitor compounds with anti-angiogenic properties remain key drugs to treat advanced renal cell carcinoma. There is a strong rationale to develop therapeutic drug monitoring for these drugs. General considerations of such monitoring of the several groups of anticancer drugs are given, with a focus on oral therapy. Pharmacokinetics and the factors of inter- and intraindividual variabilities of these tyrosine kinase inhibitors are described together with an exhaustive presentation of their pharmacokinetic/pharmacodynamic relationships. The latter was observed in studies where every patient was treated with the same dose, and the results of several prospective studies based on dose individualization support the practice of increasing individual dosage in case of low observed plasma drug concentrations. Finally, the benefits and limits of therapeutic drug monitoring as a routine practice are discussed.
Collapse
Affiliation(s)
- Florent Puisset
- Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse–Oncopole, 31059 Toulouse, France
- CRCT, Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, 31037 Toulouse, France
| | - Mourad Mseddi
- Department of Pharmacokinetics and Pharmacochemistry, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, CARPEM, 75014 Paris, France
| | - Loïc Mourey
- Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse–Oncopole, 31059 Toulouse, France
| | - Damien Pouessel
- Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse–Oncopole, 31059 Toulouse, France
| | - Benoit Blanchet
- Department of Pharmacokinetics and Pharmacochemistry, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, CARPEM, 75014 Paris, France
- UMR8038 CNRS, U1268 INSERM, Faculté de Pharmacie, Université Paris Cité, PRES Sorbonne Paris Cité, CARPEM, 75006 Paris, France
| | - Etienne Chatelut
- Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse–Oncopole, 31059 Toulouse, France
- CRCT, Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, 31037 Toulouse, France
- Correspondence: ; Tel.: +33-5-3115-5250
| | - Christine Chevreau
- Institut Claudius-Regaud, Institut Universitaire du Cancer de Toulouse–Oncopole, 31059 Toulouse, France
| |
Collapse
|
31
|
High-Dose Intermittent Treatment with the Multikinase Inhibitor Sunitinib Leads to High Intra-Tumor Drug Exposure in Patients with Advanced Solid Tumors. Cancers (Basel) 2022; 14:cancers14246061. [PMID: 36551546 PMCID: PMC9775433 DOI: 10.3390/cancers14246061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Patients with advanced cancer refractory to standard treatment were treated with sunitinib at a dose of 300 mg once every week (Q1W) or 700 mg once every two weeks (Q2W). Tumor, skin and plasma concentrations were measured and immunohistochemical staining for tumor cell proliferation (TCP), microvessel density (MVD) and T-cell infiltration was performed on tumor biopsies before and after 17 days of treatment. Oral administration of 300 mg sunitinib Q1W or 700 mg Q2W resulted in 19-fold (range 5-35×) and 37-fold higher (range 10-88×) tumor drug concentrations compared to parallel maximum plasma drug concentrations, respectively. Patients with higher tumor sunitinib concentrations had favorable progression-free and overall survival than those with lower concentrations (p = 0.046 and 0.024, respectively). In addition, immunohistochemistry of tumor biopsies revealed an induction of T-cell infiltration upon treatment. These findings provide pharmacological and biological insights in the clinical benefit from high-dose intermittent sunitinib treatment. It emphasizes the potential benefit from reaching higher tumor drug concentrations and the value of measuring TKI tumor- over plasma-concentrations. The finding that reaching higher tumor drug concentrations provides most clinical benefit in patients with treatment refractory malignancies indicates that the inhibitory potency of sunitinib may be enforced by a high-dose intermittent treatment schedule. These results provide proof of concept for testing other clinically available multitargeted tyrosine kinase inhibitors in a high-dose intermittent treatment schedule.
Collapse
|
32
|
Sheppard‐Olivares S, Bello NM, Johannes CM, Hocker SE, Biller B, Husbands B, Snyder E, McMillan M, McKee T, Wouda RM. Toceranib phosphate in the management of canine insulinoma: A retrospective multicentre study of 30 cases (2009-2019). Vet Rec Open 2022; 9:e27. [PMID: 35079406 PMCID: PMC8776903 DOI: 10.1002/vro2.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/28/2021] [Accepted: 11/22/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Insulinomas are the most common tumour of the endocrine pancreas in dogs. These malignant tumours have a high metastatic rate and limited chemotherapeutic options. The multi-receptor tyrosine kinase inhibitor sunitinib malate has benefit in the treatment of metastatic insulinoma in people. Toceranib phosphate, an analogous veterinary agent, may provide benefit for dogs. METHODS A retrospective study describing the extent and duration of clinical outcomes and adverse events (AEs) in dogs diagnosed with insulinoma and receiving toceranib. RESULTS Records for 30 dogs diagnosed with insulinoma and having received toceranib were identified from a medical record search of five university and eight referral hospitals. The median progression-free interval and overall survival time were 561 days (95% confidence interval (CI): [246, 727 days]) and 656 days (95% CI: [310, 1045 days]), respectively. Of the dogs for which the canine Response evaluation criteria for solid tumours tool could be applied, the majority (66.7%) showed either a complete response, partial response or stable disease. Time to clinical progression was associated with prior intervention and type of veterinary practice. Larger dogs were at increased risk for disease progression and death. No novel AEs were reported. CONCLUSIONS Most dogs diagnosed with insulinoma and receiving toceranib appeared to have a clinical benefit. Randomised, prospective studies are needed to better elucidate and objectively quantify the potential effect and survival benefit of toceranib therapy for management of insulinoma in dogs.
Collapse
Affiliation(s)
- Sabina Sheppard‐Olivares
- Department of Clinical SciencesCollege of Veterinary MedicineKansas State UniversityManhattanKansasUSA
- Present address:
3901 Guadalupe Street, Austin, TX 78751, USA.
| | - Nora M. Bello
- Department of StatisticsCollege of Arts and SciencesKansas State UniversityManhattanKansasUSA
| | - Chad M. Johannes
- Department of Clinical SciencesCollege of Veterinary MedicineIowa State UniversityAmesIowaUSA
| | - Samuel E. Hocker
- Ontario Veterinary CollegeUniversity of GuelphGuelphOntarioCanada
- Present address:
Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS 66506, USA.
| | - Barbara Biller
- Flint Animal Cancer CenterCollege of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
- Present address:
4120 Clydesdale Pkwy, Loveland, CO 80538, USA.
| | - Brian Husbands
- Veterinary Clinical Sciences DepartmentCollege of Veterinary MedicineUniversity of MinnesotaSt. PaulMinnesotaUSA
- Present address:
Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 1900 Coffey Road, Columbus, OH 43210, USA.
| | - Elizabeth Snyder
- Department of Medical SciencesSchool of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Present address:
BluePearl Specialty and Emergency Pet Hospital, 1646 Spring Cypress Rd Ste 100, Spring, TX 77388, USA.
| | - Mattison McMillan
- Las Vegas Veterinary Specialty CenterLas VegasNevadaUSA
- Present address:
College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.
| | - Talon McKee
- Clinical Studies DepartmentVCA Inc.Los AngelesCaliforniaUSA
| | - Raelene M. Wouda
- Department of Clinical SciencesCollege of Veterinary MedicineKansas State UniversityManhattanKansasUSA
| |
Collapse
|
33
|
Muhandiramge J, Zalcberg JR, van Londen GJ, Warner ET, Carr PR, Haydon A, Orchard SG. Cardiovascular Disease in Adult Cancer Survivors: a Review of Current Evidence, Strategies for Prevention and Management, and Future Directions for Cardio-oncology. Curr Oncol Rep 2022; 24:1579-1592. [PMID: 35796941 PMCID: PMC9606033 DOI: 10.1007/s11912-022-01309-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Cardiovascular disease is long-term complication of both cancer and anti-cancer treatment and can have significant ramifications for health-related quality of life and mortality. This narrative review explores the current evidence linking cardiovascular disease and cancer, as well as exploring strategies for the prevention and management of cardiovascular disease, and outlines future opportunities in the field of cardio-oncology. RECENT FINDINGS Cancer confers risk for various cardiovascular diseases including heart failure, cardiomyopathy, arrhythmia, coronary heart disease, stroke, venous thromboembolism, and valvular heart disease. Cancer treatment, in particular agents such as platinum-based chemotherapy, anthracyclines, hormonal treatments, and thoracic radiotherapy, further increases risk. While cardiovascular disease can be identified early and effectively managed in cancer survivors, cardiovascular screening and management does not typically feature in routine long-term cancer care of adult cancer survivors. Cancer and cancer treatment can accelerate the development of cardiovascular disease. Further research into screening and management strategies for cardiovascular disease, along with evidence-based guidelines, is required to ensure adult cancer survivors receive appropriate long-term care.
Collapse
Affiliation(s)
- Jaidyn Muhandiramge
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia.
- Austin Health, Heidelberg, VIC, Australia.
| | - John R Zalcberg
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia
- Department of Medical Oncology, Alfred Hospital, Melbourne, VIC, Australia
| | - G J van Londen
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Erica T Warner
- Clinical and Translational Epidemiology Unit, MGH Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Prudence R Carr
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia
| | - Andrew Haydon
- Department of Medical Oncology, Alfred Hospital, Melbourne, VIC, Australia
| | - Suzanne G Orchard
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia
| |
Collapse
|
34
|
Addressing the Role of Angiogenesis in Patients with Advanced Pancreatic Neuroendocrine Tumors Treated with Everolimus: A Biological Prospective Analysis of Soluble Biomarkers and Clinical Outcomes. Cancers (Basel) 2022; 14:cancers14184471. [PMID: 36139632 PMCID: PMC9497075 DOI: 10.3390/cancers14184471] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Background: The success of targeted therapies in the treatment of pancreatic neuroendocrine tumors has emphasized the strategy of targeting angiogenesis and the PI3K/AKT/mTOR pathway. However, the major challenge in the targeted era remains the early identification of resistant tumors especially when the efficacy is rarely associated to a clear tumor shrinkage at by imaging assessment. Methods: In this prospective study (NCT02305810) we investigated the predictive and prognostic role of soluble biomarkers of angiogenesis turnover (VEGF, bFGF, VEGFR2, TSP-1) circulating endothelial cells and progenitors, in 43 patients with metastatic panNET receiving everolimus. Results: Among all tested biomarkers, we found a specific subpopulation of circulating cells, CD31+CD140b-, with a significantly increased tumor progression hazard for values less or equal to the first quartile. Conclusion: Our study suggested the evidence that circulating cells might be surrogate biomarkers of angiogenesis activity in patients treated with everolimus and their baseline levels can be correlated with survival. However, further studies are now needed to validate the role of these cells as surrogate markers for the selection of patients to be candidates for antiangiogenic treatments.
Collapse
|
35
|
Ma C, Wu Z, Wang X, Huang M, Wei X, Wang W, Qu H, Qiaolongbatu X, Lou Y, Jing L, Fan G. A systematic comparison of anti-angiogenesis efficacy and cardiotoxicity of receptor tyrosine kinase inhibitors in zebrafish model. Toxicol Appl Pharmacol 2022; 450:116162. [PMID: 35830948 DOI: 10.1016/j.taap.2022.116162] [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: 03/09/2022] [Revised: 07/01/2022] [Accepted: 07/07/2022] [Indexed: 10/17/2022]
Abstract
Pathological angiogenesis is fundamental to progression of cancerous tumors and blinding eye diseases. Anti-angiogenic receptor tyrosine kinase inhibitors (TKIs) are in broad use for the treatment of these diseases. With more and more TKIs available, it is a challenge to make an optimal choice. It remains unclear whether TKIs demonstrate similar anti-angiogenesis activities in different tissues. Many TKIs have shown varying degrees of toxic effects that should also be considered in clinical use. This study investigates the anti-angiogenic effects of 13 FDA-approved TKIs on the intersegmental vessels (ISVs), subintestinal vessels (SIVs) and retinal vasculature in zebrafish embryos. The results show that vascular endothelial growth factor receptor TKIs (VEGFR-TKIs) exhibit anti-angiogenic abilities similarly on ISVs and SIVs, and their efficacy is consistent with their IC50 values against VEGFR2. In addition, VEGFR-TKIs selectively induces the apoptosis of endothelial cells in immature vessels. Among all TKIs tested, axitinib demonstrates a strong inhibition on retinal neovascularization at a low dose that do not strongly affect ISVs and SIVs, supporting its potential application for retinal diseases. Zebrafish embryos demonstrate cardiotoxicity after VEGFR-TKIs treatment, and ponatinib and sorafenib show a narrow therapeutic window, suggesting that these two drugs may need to be dosed more carefully in patients. We propose that zebrafish is an ideal model for studying in vivo antiangiogenic efficacy and cardiotoxicity of TKIs.
Collapse
Affiliation(s)
- Cui Ma
- School of Pharmacy, Shanghai Jiao Tong University, Building 6-312, Shanghai 200240, PR China; Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Zhenghua Wu
- School of Pharmacy, Shanghai Jiao Tong University, Building 6-312, Shanghai 200240, PR China; Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Xue Wang
- School of Pharmacy, Shanghai Jiao Tong University, Building 6-312, Shanghai 200240, PR China
| | - Mengling Huang
- School of Pharmacy, Shanghai Jiao Tong University, Building 6-312, Shanghai 200240, PR China
| | - Xiaona Wei
- School of Pharmacy, Shanghai Jiao Tong University, Building 6-312, Shanghai 200240, PR China
| | - Wei Wang
- School of Pharmacy, Shanghai Jiao Tong University, Building 6-312, Shanghai 200240, PR China
| | - Han Qu
- School of Pharmacy, Shanghai Jiao Tong University, Building 6-312, Shanghai 200240, PR China; Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Xijier Qiaolongbatu
- School of Pharmacy, Shanghai Jiao Tong University, Building 6-312, Shanghai 200240, PR China; Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Yuefen Lou
- Department of Pharmacy, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200434, PR China.
| | - Lili Jing
- School of Pharmacy, Shanghai Jiao Tong University, Building 6-312, Shanghai 200240, PR China.
| | - Guorong Fan
- School of Pharmacy, Shanghai Jiao Tong University, Building 6-312, Shanghai 200240, PR China; Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China.
| |
Collapse
|
36
|
Modica R, Liccardi A, Minotta R, Cannavale G, Benevento E, Colao A. Therapeutic strategies for patients with neuroendocrine neoplasms: current perspectives. Expert Rev Endocrinol Metab 2022; 17:389-403. [PMID: 35822906 DOI: 10.1080/17446651.2022.2099840] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
INTRODUCTION Neuroendocrine neoplasms (NENs) are a heterogeneous group of malignancies mainly arising in the gastroenteropancreatic (GEP) and bronchopulmonary systems, with steadily increasing incidence. The therapeutic landscape has widened and the therapeutic strategy should be based on new sequences and combinations, still debated. AREAS COVERED Herein, we provide an overview of current approved pharmacological treatments in patients with NENs, with the aim to summarize evidence of efficacy of the main different options in GEP and pulmonary NENs, principally focusing on somatostatin analogs (SSAs), targeted therapy with everolimus and sunitinib, peptide receptor radionuclide therapy (PRRT) and chemotherapy. We discuss biological rationale and toxicities, including current indications according to differentiation and placement in the therapeutic algorithm, clinical trials, and combinations. Furthermore, we recommend areas for further research. EXPERT OPINION Therapeutic management of patients with NENs represents a challenge for clinicians and the identification of effective sequences and combinations is of utmost importance. Major efforts should be directed to early identify and overcome resistance and to limit toxicity. The progress in the therapeutic management of NENs grows faster and the choice of the best approach should be based on randomized clinical trials, as well as on long-term, real-world data.
Collapse
Affiliation(s)
- Roberta Modica
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - Alessia Liccardi
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - Roberto Minotta
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - Giuseppe Cannavale
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - Elio Benevento
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - Annamaria Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
- UNESCO Chair, Education for Health and Sustainable Development, Federico II University, Naples, Italy
| |
Collapse
|
37
|
Targeting Tyrosine Kinases in Ovarian Cancer: Small Molecule Inhibitor and Monoclonal Antibody, Where Are We Now? Biomedicines 2022; 10:biomedicines10092113. [PMID: 36140214 PMCID: PMC9495728 DOI: 10.3390/biomedicines10092113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 12/27/2022] Open
Abstract
Ovarian cancer is one of the most lethal gynaecological malignancies worldwide. Despite high success rates following first time treatment, this heterogenous disease is prone to recurrence. Oncogenic activity of receptor tyrosine kinases is believed to drive the progression of ovarian cancer. Here we provide an update on the progress of the therapeutic targeting of receptor tyrosine kinases in ovarian cancer. Broadly, drug classes that inhibit tyrosine kinase/pathways can be classified as small molecule inhibitors, monoclonal antibodies, or immunotherapeutic vaccines. Small molecule inhibitors tested in clinical trials thus far include sorafenib, sunitinib, pazopanib, tivantinib, and erlotinib. Monoclonal antibodies include bevacizumab, cetuximab, pertuzumab, trastuzumab, and seribantumab. While numerous trials have been carried out, the results of monotherapeutic agents have not been satisfactory. For combination with chemotherapy, the monoclonal antibodies appear more effective, though the efficacy is limited by low frequency of target alteration and a lack of useful predictive markers for treatment stratification. There remain critical gaps for the treatment of platinum-resistant ovarian cancers; however, platinum-sensitive tumours may benefit from the combination of tyrosine kinase targeting drugs and PARP inhibitors. Immunotherapeutics such as a peptide B-cell epitope vaccine and plasmid-based DNA vaccine have shown some efficacy both as monotherapeutic agents and in combination therapy, but require further development to validate current findings. In conclusion, the tyrosine kinases remain attractive targets for treating ovarian cancers. Future development will need to consider effective drug combination, frequency of target, and developing predictive biomarker.
Collapse
|
38
|
Abudayah A, Daoud S, Al-Sha'er M, Taha M. Pharmacophore Modeling of Targets Infested with Activity Cliffs via Molecular Dynamics Simulation Coupled with QSAR and Comparison with other Pharmacophore Generation Methods: KDR as Case Study. Mol Inform 2022; 41:e2200049. [PMID: 35973966 DOI: 10.1002/minf.202200049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 08/15/2022] [Indexed: 11/07/2022]
Abstract
Activity cliffs (ACs) are defined as pairs of structurally similar compounds with large difference in their potencies against certain biotarget. We recently proposed that potent AC members induce significant entropically-driven conformational modifications of the target that unveil additional binding interactions, while their weakly-potent counterparts are enthalpically-driven binders with little influence on the protein target. We herein propose to extract pharmacophores for ACs-infested target(s) from molecular dynamics (MD) frames of purely "enthalpic" potent binder(s) complexed within the particular target. Genetic function algorithm/machine learning (GFA/ML) can then be employed to search for the best possible combination of MD pharmacophore(s) capable of explaining bioactivity variations within a list of inhibitors. We compared the performance of this approach with established ligand-based and structure-based methods. Kinase inserts domain receptor (KDR) was used as a case study. KDR plays a crucial role in angiogenic signaling and its inhibitors have been approved in cancer treatment. Interestingly, GFA/ML selected, MD-based, pharmacophores were of comparable performances to ligand-based and structure-based pharmacophores. The resulting pharmacophores and QSAR models were used to capture hits from the national cancer institute list of compounds. The most active hit showed anti-KDR IC50 of 2.76 µM.
Collapse
Affiliation(s)
| | | | | | - Mutasem Taha
- Faculty of pharmacy,University of jordan, JORDAN
| |
Collapse
|
39
|
Noda S, Morita SY, Terada T. Dose Individualization of Oral Multi-Kinase Inhibitors for the Implementation of Therapeutic Drug Monitoring. Biol Pharm Bull 2022; 45:814-823. [PMID: 35786588 DOI: 10.1248/bpb.b21-01098] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Oral multi-kinase inhibitors have transformed the treatment landscape for various cancer types and provided significant improvements in clinical outcomes. These agents are mainly approved at fixed doses, but the large inter-individual variability in pharmacokinetics and pharmacodynamics (efficacy and safety) has been an unsolved clinical issue. For example, certain patients treated with oral multi-kinase inhibitors at standard doses have severe adverse effects and require dose reduction and discontinuation, yet other patients have a suboptimal response to these drugs. Consequently, optimizing the dosing of oral multi-kinase inhibitors is important to prevent over-dosing or under-dosing. To date, multiple studies on the exposure-efficacy/toxicity relationship of molecular targeted therapy have been attempted for the implementation of therapeutic drug monitoring (TDM) strategies. In this milieu, we recently conducted research on several multi-kinase inhibitors, such as sunitinib, pazopanib, sorafenib, and lenvatinib, with the aim to optimize their treatment efficacy using a pharmacokinetic/pharmacodynamic approach. Among them, sunitinib use is an example of successful TDM implementation. Sunitinib demonstrated a significant correlation between drug exposure and treatment efficacy or toxicities. As a result, TDM services for sunitinib has been covered by the National Health Insurance program in Japan since April 2018. Additionally, other multi-kinase targeted anticancer drugs have promising data regarding the exposure-efficacy/toxicity relationship, suggesting the possibility of personalization of drug dosage. In this review, we provide a comprehensive summary of the clinical evidence for dose individualization of multi-kinase inhibitors and discuss the utility of TDM of multi-kinase inhibitors, especially sunitinib, pazopanib, sorafenib, and lenvatinib.
Collapse
Affiliation(s)
- Satoshi Noda
- Department of Pharmacy, Shiga University of Medical Science Hospital
| | - Shin-Ya Morita
- Department of Pharmacy, Shiga University of Medical Science Hospital
| | - Tomohiro Terada
- Department of Pharmacy, Shiga University of Medical Science Hospital.,Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital
| |
Collapse
|
40
|
Jiang X, Xiong F, Fu Q, Peng H, Jing Y, Rexiti K, Wei X, Tao S. Hematologic toxicities of sunitinib in patients with gastrointestinal stromal tumors: a systematic review and meta-analysis. Int J Colorectal Dis 2022; 37:1525-1534. [PMID: 35780257 DOI: 10.1007/s00384-022-04214-7] [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] [Accepted: 06/27/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE Sunitinib offers a significant survival benefit to patients with imatinib-resistant gastrointestinal stromal tumors (GIST). However, the incidence and risk of sunitinib-induced hematologic toxicities in such a population are often overlooked and have not been well characterized. This meta-analysis was performed to assess the summary incidence and risk of hematologic toxicities secondary to sunitinib in patients with GIST. METHODS Searches were performed in PubMed, Embase, Cochrane Library, and Web of Science as well as ClinicalTrials.gov to identify relevant studies up to April 2022. Studies with adequate safety profile, including anemia, neutropenia, and thrombocytopenia, were included to calculate the pooled incidence, relative risk (RR), and corresponding 95% confidence intervals (CIs). This study was registered with PROSPERO under number CRD42022328202. RESULTS A total of 2593 patients from 13 studies were included in the present meta-analysis. For patients with GIST assigned to sunitinib, the overall incidences of all-grade anemia, neutropenia, and thrombocytopenia were 26.2% (95% CI, 14.9-39.4%), 41.8% (95% CI, 29.0-55.1%), and 36.4% (95% CI, 22.8-51.1%), respectively. Regarding high-grade (grades 3 and 4) events, there were 4.7% (95% CI, 3.8-5.6%) for anemia, 9.3% (95% CI, 5.6-13.7%) for neutropenia and 5.0% (95% CI, 2.9-7.3%) for thrombocytopenia. Compared to placebo arms, sunitinib was related to an increased risk of high-grade neutropenia with an RR of 10.39 (95% CI, 1.53-70.72; p = 0.017). CONCLUSIONS Sunitinib carries a relatively high incidence of hematologic toxicities and a substantial increased risk of high-grade neutropenia in patients with GIST. Appropriate prevention and management seem to be inevitable.
Collapse
Affiliation(s)
- Xuehui Jiang
- School of Pharmacy, Nanchang University, Nanchang, China.,Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fangfang Xiong
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Qun Fu
- School of Pharmacy, Nanchang University, Nanchang, China.,Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongwei Peng
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan Jing
- Department of Pharmacy, Linyi Central Hospital, Linyi, China
| | - Kaisaner Rexiti
- School of Pharmacy, Nanchang University, Nanchang, China.,Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaohua Wei
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Song Tao
- Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
41
|
Burnham EA, Abouda AA, Bissada JE, Nardone-White DT, Beers JL, Lee J, Vergne MJ, Jackson KD. Interindividual Variability in Cytochrome P450 3A and 1A Activity Influences Sunitinib Metabolism and Bioactivation. Chem Res Toxicol 2022; 35:792-806. [PMID: 35484684 PMCID: PMC9131896 DOI: 10.1021/acs.chemrestox.1c00426] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Sunitinib is an orally administered tyrosine kinase inhibitor associated with idiosyncratic hepatotoxicity; however, the mechanisms of this toxicity remain unclear. We have previously shown that cytochromes P450 1A2 and 3A4 catalyze sunitinib metabolic activation via oxidative defluorination leading to a chemically reactive, potentially toxic quinoneimine, trapped as a glutathione (GSH) conjugate (M5). The goals of this study were to determine the impact of interindividual variability in P450 1A and 3A activity on sunitinib bioactivation to the reactive quinoneimine and sunitinib N-dealkylation to the primary active metabolite N-desethylsunitinib (M1). Experiments were conducted in vitro using single-donor human liver microsomes and human hepatocytes. Relative sunitinib metabolite levels were measured by liquid chromatography-tandem mass spectrometry. In human liver microsomes, the P450 3A inhibitor ketoconazole significantly reduced M1 formation compared to the control. The P450 1A2 inhibitor furafylline significantly reduced defluorosunitinib (M3) and M5 formation compared to the control but had minimal effect on M1. In CYP3A5-genotyped human liver microsomes from 12 individual donors, M1 formation was highly correlated with P450 3A activity measured by midazolam 1'-hydroxylation, and M3 and M5 formation was correlated with P450 1A2 activity estimated by phenacetin O-deethylation. M3 and M5 formation was also associated with P450 3A5-selective activity. In sandwich-cultured human hepatocytes, the P450 3A inducer rifampicin significantly increased M1 levels. P450 1A induction by omeprazole markedly increased M3 formation and the generation of a quinoneimine-cysteine conjugate (M6) identified as a downstream metabolite of M5. The nonselective P450 inhibitor 1-aminobenzotriazole reduced each of these metabolites (M1, M3, and M6). Collectively, these findings indicate that P450 3A activity is a key determinant of sunitinib N-dealkylation to the active metabolite M1, and P450 1A (and potentially 3A5) activity influences sunitinib bioactivation to the reactive quinoneimine metabolite. Accordingly, modulation of P450 activity due to genetic and/or nongenetic factors may impact the risk of sunitinib-associated toxicities.
Collapse
Affiliation(s)
- Elizabeth A Burnham
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204, United States
| | - Arsany A Abouda
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204, United States
| | - Jennifer E Bissada
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204, United States
| | - Dasean T Nardone-White
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Jessica L Beers
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Jonghwa Lee
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Matthew J Vergne
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204, United States
| | - Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
42
|
Gandhi KA, Joshi A, Mehta P, Gurjar M, Rane P, Sharma J, Patil A, Nookala M, Noronha V, Prabhash K, Gota V. Feasibility of therapeutic drug monitoring of sunitinib and its implications on response and toxicity in patients with metastatic renal cell cancer. Cancer Chemother Pharmacol 2022; 89:751-759. [PMID: 35441269 DOI: 10.1007/s00280-022-04432-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/02/2022] [Indexed: 01/05/2023]
Abstract
PURPOSE Sunitinib is an oral tyrosine kinase inhibitor approved for the treatment of metastatic renal cell carcinoma (mRCC). High variability in pharmacokinetics coupled with a proven exposure-effect relationship makes sunitinib an ideal candidate for therapeutic drug monitoring (TDM). The feasibility of TDM of sunitinib in patients with mRCC was evaluated in this prospective observational study in a real-world scenario. METHODS Seventy patients with mRCC treated with sunitinib at a fixed dose of 50 mg per day were enrolled in the study. Total trough plasma level (TTL) of sunitinib (sunitinib and its active metabolite, SU12662), was measured between days 14/15 of cycle 1. The discriminatory potential of TTL of sunitinib for the prediction of responders and occurrence of grade ≥ 3 toxicity was determined using receiver operating characteristic (ROC) curve. RESULTS The median TTL of sunitinib was 76 ng/mL. Forty six out of 70 patients were evaluable for response, whereas 60 out of 70 patients were evaluable for toxicity. Threshold concentrations obtained from ROC analysis showed that TTL of 60.75 ng/mL and 82.3 ng/mL was discriminatory for response and occurrence of grade ≥ 3 toxicity respectively. 31/34 (91.7%) patients having TTL ≥ 60.75 ng/mL responded to treatment, while only 5/12 (41.6%) responded when TTL was < 60.75 ng/mL (P = 0.001). On the other hand, the incidence of grade ≥ 3 toxicity was 9/24 (37.7%) in patients with TTL ≥ 82.3 ng/mL compared to 4/36 (11.1%) in patients with TTL < 82.3 ng/mL (P = 0.024). CONCLUSION The TTL range of 60.75-82.3 ng/mL was found to be optimal in terms of safety and efficacy. More than 50% of patients in our cohort attained TTL of sunitinib outside the optimal range, thus demonstrating the feasibility of TDM to improve safety and efficacy of sunitinib in mRCC.
Collapse
Affiliation(s)
- Khushboo A Gandhi
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Sector-22, Kharghar, Navi Mumbai, 410210, India
| | - Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai, 400012, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Parsshava Mehta
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Sector-22, Kharghar, Navi Mumbai, 410210, India
| | - Murari Gurjar
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Sector-22, Kharghar, Navi Mumbai, 410210, India
| | - Pallavi Rane
- Department of Biostatistics, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, 410210, India
| | - Jyoti Sharma
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Sector-22, Kharghar, Navi Mumbai, 410210, India
| | - Anand Patil
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Sector-22, Kharghar, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Manjunath Nookala
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Sector-22, Kharghar, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai, 400012, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai, 400012, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Vikram Gota
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Sector-22, Kharghar, Navi Mumbai, 410210, India.
- Homi Bhabha National Institute, Mumbai, 400094, India.
| |
Collapse
|
43
|
Don't Give Up Too Soon! Management of Metastatic Duodenal GISTs with Tumor-Bowel Fistula: A Case Report and Literature Review. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2022. [DOI: 10.1016/j.cpccr.2022.100165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
44
|
Chen T, Chen J, Chen C, Guo J, He X, Zheng S, Liu M, Zheng B. Systematic review and cost-effectiveness of pharmacokinetically guided sunitinib individualized treatment for patients with metastatic renal cell carcinoma. Ther Adv Med Oncol 2022; 14:17588359221085212. [PMID: 35371296 PMCID: PMC8972915 DOI: 10.1177/17588359221085212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/15/2022] [Indexed: 11/21/2022] Open
Abstract
Background: Sunitinib has a narrow therapeutic window, with considerable differences between patients. Dosing based on pharmacokinetics (PK) may help overcome some of those issues. This study aims to evaluate and compare the cost-effectiveness of PK-guided individualized treatment of sunitinib with its standard dose in patients with metastatic renal cell carcinoma (mRCC). Methods: A comprehensive literature search was performed, and relevant values were used to provide information for the decision analysis model. Utility data were derived from published studies, and costs were obtained from the perspective of payers in China and the United States. A Markov model was established to evaluate the associated costs and health outcomes for patients. The primary outputs of the model included lifetime costs, quality-adjusted life years (QALYs), and incremental cost-effectiveness ratio (ICER). One-way and probability sensitivity analyses were conducted to evaluate the potential uncertainties of parameters. Results: Cost-effective analysis showed that the QALY of the PK-guided group increased by 0.83 compared with that in the standard dose group. From the perspective of both countries’ health systems, the cost of PK-guided dose was lower than that of standard dose. Hence, PK-guided treatment was the dominant strategy. One-way and probability sensitivity analyses confirmed the reliability of these results. Conclusion: On the basis of currently available data, PK-guided sunitinib treatment may be a safe, effective, and economical intervention for patients with mRCC.
Collapse
Affiliation(s)
- Tingting Chen
- College of Pharmacy, Fujian Medical University, Fuzhou, ChinaDepartment of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiahe Chen
- Department of Pharmaceutical and Health Economics, School of Pharmacy, University of Southern California, Los Angeles, CA, USALeonard D. Schaeffer Center for Health Policy & Economics, University of Southern California, Los Angeles, CA, USA
| | - Chaoxin Chen
- College of Pharmacy, Fujian Medical University, Fuzhou, ChinaDepartment of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianming Guo
- College of Pharmacy, Fujian Medical University, Fuzhou, ChinaDepartment of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xin He
- College of Pharmacy, Fujian Medical University, Fuzhou, ChinaDepartment of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Song Zheng
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Maobai Liu
- Fujian Medical University Union Hospital, 29,Xinquan Road, Gulou District, Fuzhou City 350001, Fujian Province, China
| | - Bin Zheng
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian, ChinaCollege of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
45
|
Krens SD, van Erp NP, Groenland SL, Moes DJAR, Mulder SF, Desar IME, van der Hulle T, Steeghs N, van Herpen CML. Exposure-response analyses of cabozantinib in patients with metastatic renal cell cancer. BMC Cancer 2022; 22:228. [PMID: 35236333 PMCID: PMC8892746 DOI: 10.1186/s12885-022-09338-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/17/2022] [Indexed: 11/10/2022] Open
Abstract
Aim In the registration trial, cabozantinib exposure ≥ 750 ng/mL correlated to improved tumor size reduction, response rate and progression free survival (PFS) in patients with metastatic renal cell cancer (mRCC). Because patients in routine care often differ from patients in clinical trials, we explored the cabozantinib exposure–response relationship in patients with mRCC treated in routine care. Methods Cabozantinib trough concentrations (Cmin) were collected and average exposure was calculated per individual. Exposure–response analyses were performed using the earlier identified target of Cmin > 750 ng/mL and median Cmin. In addition, the effect of dose reductions on response was explored. PFS was used as measure of response. Results In total, 59 patients were included:10% were classified as favourable, 61% as intermediate and 29% as poor IMDC risk group, respectively. Median number of prior treatment lines was 2 (0–5). Starting dose was 60 mg in 46%, 40 mg in 42% and 20 mg in 12% of patients. Dose reductions were needed in 58% of patients. Median Cmin was 572 ng/mL (IQR: 496–701). Only 17% of patients had an average Cmin ≥ 750 ng/mL. Median PFS was 52 weeks (95% CI: 40–64). No improved PFS was observed for patients with Cmin ≥ 750 ng/mL or ≥ 572 ng/ml. A longer PFS was observed for patients with a dose reduction vs. those without (65 vs. 31 weeks, p = .001). After incorporating known covariates (IMDC risk group and prior treatment lines (< 2 vs. ≥ 2)) in the multivariable analysis, the need for dose reduction remained significantly associated with improved PFS (HR 0.32, 95% CI:0.14–0.70, p = .004). Conclusion In these explorative analyses, no clear relationship between increased cabozantinib exposure and improved PFS was observed. Average cabozantinib exposure was below the previously proposed target in 83% of patients. Future studies should focus on validating the cabozantinib exposure required for long term efficacy. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09338-1.
Collapse
Affiliation(s)
- Stefanie D Krens
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.
| | - Nielka P van Erp
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Stefanie L Groenland
- Division of Medical Oncology, Department of Clinical Pharmacology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Sasja F Mulder
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Tom van der Hulle
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Neeltje Steeghs
- Division of Medical Oncology, Department of Clinical Pharmacology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Carla M L van Herpen
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
46
|
Vorolanib, a novel tyrosine receptor kinase receptor inhibitor with potent preclinical anti-angiogenic and anti-tumor activity. Mol Ther Oncolytics 2022; 24:577-584. [PMID: 35252556 PMCID: PMC8861424 DOI: 10.1016/j.omto.2022.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 01/07/2022] [Indexed: 01/13/2023] Open
Abstract
Vorolanib (CM082) is a multi-targeted tyrosine kinase receptor inhibitor with a short half-life and limited tissue accumulation that has been shown to reduce choroidal neovascularization in rats. In this preclinical study, vorolanib demonstrated competitive binding and inhibitory activities with KDR, PDGFRβ, FLT3, and C-Kit, and inhibited RET and AMPKα1 more weakly than sunitinib, indicating more stringent kinase selectivity. Vorolanib inhibited vascular endothelial growth factor (VEGF)-induced proliferation of human umbilical vein endothelial cells (HUVECs) and HUVEC tube formation in vitro. In mouse xenograft models, vorolanib inhibited tumor growth of MV-4-11, A549, 786-O, HT-29, BxPC-3, and A375 cells in a dose-dependent fashion. Complete tumor regression was achieved in the MV-4-11 xenograft model. No significant toxicities were observed in vorolanib groups, whereas a significant negative impact on body weights was observed in the sunitinib group at a dose of 40 mg/kg qd. Overall, vorolanib is a novel multi-kinase receptor inhibitor with potent preclinical anti-angiogenic and anti-tumor activity that is potentially less toxic than other similar kinase inhibitors.
Collapse
|
47
|
Ferrer F, Chauvin J, Deville JL, Ciccolini J. Adaptive dosing of sunitinib in a metastatic renal cell carcinoma patient: when in silico modeling helps to go quicker to the point. Cancer Chemother Pharmacol 2022; 89:565-569. [PMID: 35147741 DOI: 10.1007/s00280-021-04383-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/07/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Adaptive dosing strategy with oral targeted therapies in oncology is mostly based upon clinical signs. Using pharmacokinetics (PK) models to customize dosing could help saving time, i.e., by predicting clinical outcome through early monitoring of drug levels. CASE REPORT We present the case of a metastatic renal cell carcinoma patient treated with standard Sunitinib dosing (i.e., 50 mg QD). Clinical signs suggested lack of efficacy. Therapeutic Drug Monitoring (TDM) confirmed that exposure was below the expected target exposure. PK modeling suggested that dosing could be increased safely to 75 mg QD. Sunitinib dosing was instead changed empirically to 62.5 mg only, increasing drug exposure to the lower part of the therapeutic window. Resolution of bone pains plus Stable Disease were observed. Even though further modeling suggested to increase Sunitinib dosing to 75 mg again, the intermediate dosing was maintained for the subsequent cycles to preserve the safety. Unfortunately, severe pains plus degradation of the general state were reported and imaging showed Progressive Disease. The patient was finally switched to alternative therapy, without being treated at the 75 mg level of Suntitinib. CONCLUSIONS AND DISCUSSION This case suggests that model-based adaptive dosing could have allowed to reach quicker the best dosing with Sunitinib, thus possibly ensuring a better management of this patient. Model-informed dosing should be used instead of empirical search for the most appropriate dosing to ensure a good benefit/risk ratio with Sunitinib, especially in the context of such aggressive disease.
Collapse
Affiliation(s)
- Florent Ferrer
- COMPO Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068 Inria Centre de Recherches Sophia Méditerranée, Aix Marseille Université, 13385, Marseille, France.,SMARTc, Centre de Recherche en Cancérologie de Marseille, Inserm U1068 Aix Marseille Université, 13385, Marseille, France.,Laboratoire de Pharmacocinétique Clinique Et de Toxicologie, La Timone University Hospital of Marseille, 13385, Marseille, France
| | | | - Jean-Laurent Deville
- Medical Oncology, La Timone University Hospital of Marseille, 13385, Marseille, France
| | - Joseph Ciccolini
- COMPO Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068 Inria Centre de Recherches Sophia Méditerranée, Aix Marseille Université, 13385, Marseille, France. .,SMARTc, Centre de Recherche en Cancérologie de Marseille, Inserm U1068 Aix Marseille Université, 13385, Marseille, France. .,Laboratoire de Pharmacocinétique Clinique Et de Toxicologie, La Timone University Hospital of Marseille, 13385, Marseille, France.
| |
Collapse
|
48
|
Franczyk B, Rysz J, Gluba-Brzózka A. Pharmacogenetics of Drugs Used in the Treatment of Cancers. Genes (Basel) 2022; 13:311. [PMID: 35205356 PMCID: PMC8871547 DOI: 10.3390/genes13020311] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
Pharmacogenomics is based on the understanding of the individual differences in drug use, the response to drug therapy (efficacy and toxicity), and the mechanisms underlying variable drug responses. The identification of DNA variants which markedly contribute to inter-individual variations in drug responses would improve the efficacy of treatments and decrease the rate of the adverse side effects of drugs. This review focuses only on the impact of polymorphisms within drug-metabolizing enzymes on drug responses. Anticancer drugs usually have a very narrow therapeutic index; therefore, it is very important to use appropriate doses in order to achieve the maximum benefits without putting the patient at risk of life-threatening toxicities. However, the adjustment of the appropriate dose is not so easy, due to the inheritance of specific polymorphisms in the genes encoding the target proteins and drug-metabolizing enzymes. This review presents just a few examples of such polymorphisms and their impact on the response to therapy.
Collapse
Affiliation(s)
| | | | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland; (B.F.); (J.R.)
| |
Collapse
|
49
|
Gao W, Zhang S, Guorong L, Liu Q, Zhu A, Gui F, Zou Y, Wu Y, Luo Y, Hong Z. Nc886 promotes renal cancer cell drug-resistance by enhancing EMT through Rock2 phosphorylation-mediated β-catenin nuclear translocation. Cell Cycle 2022; 21:340-351. [PMID: 34974812 PMCID: PMC8855853 DOI: 10.1080/15384101.2021.2020431] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Drug resistance is a significant challenge in the present treatment regimens of renal cell carcinoma (RCC). Our previous study confirmed that nc886 functions as an oncogene in RCC. Nevertheless, the role and underlying mechanism of nc886 in RCC drug resistance are unclear. In the present study, Sunitinib and Everolimus treatment, respectively, downregulated nc886 expression in a dose-dependent manner in all four renal cancer cell lines. Nc886 overexpression in 786-O cells and ACHN cells significantly reduced the sensitivity of cancer cells to both Sunitinib and Everolimus treatment, respectively, by promoting cell viability and inhibiting cell apoptosis, whereas nc886 silencing increased cancer cell sensitivity. In renal cancer cell line with the highest drug-resistance, 786-O cells, Sunitinib, or Everolimus treatment enhanced the cellular EMT and was further enhanced by nc886 overexpression while attenuated by nc886 silencing. In 786-O cells, nc886 overexpression significantly promoted EMT, ROCK2 phosphorylation, and β-catenin nucleus translocation under Sunitinib or Everolimus treatment. Moreover, ROCK2 silencing significantly reversed the effects of nc886 overexpression on EMT, ROCK2 phosphorylation, and β-catenin nucleus translocation, as well as drug-resistant renal cancer cell viability and apoptosis. In conclusion, it was demonstrated that nc886 promotes renal cancer cell proliferation, migration, and invasion, as demonstrated previously. nc886 also promotes renal cancer cell drug-resistance to Sunitinib or Everolimus by promoting EMT through Rock2 phosphorylation-mediated nuclear translocation of β-catenin.
Collapse
Affiliation(s)
- Weiyin Gao
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Shouhua Zhang
- Department of General Surgery, Jiangxi Children’s Hospital, Nanchang, Jiangxi Province, China
| | - Li Guorong
- Department of Urology, North Hospital, Chu Saint-Etienne, University of Jean-Monnet, Saint-Etienne, France
| | - Queling Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Anyi Zhu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Fu Gui
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yan Zou
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yiguo Wu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yang Luo
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhengdong Hong
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China,CONTACT Zhengdong Hong The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Nanchang, Jiangxi Province, China
| |
Collapse
|
50
|
Xiong Y, Liu T, Chen T, Hansen J, Hu B, Chen Y, Jayaraman G, Schürer S, Vidovic D, Goldfarb J, Sobie EA, Birtwistle MR, Iyengar R, Li H, Azeloglu EU. Proteomic cellular signatures of kinase inhibitor-induced cardiotoxicity. Sci Data 2022; 9:18. [PMID: 35058449 PMCID: PMC8776854 DOI: 10.1038/s41597-021-01114-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 12/14/2021] [Indexed: 11/09/2022] Open
Abstract
Drug Toxicity Signature Generation Center (DToxS) at the Icahn School of Medicine at Mount Sinai is one of the centers for the NIH Library of Integrated Network-Based Cellular Signatures (LINCS) program. Its key aim is to generate proteomic and transcriptomic signatures that can predict cardiotoxic adverse effects of kinase inhibitors approved by the Food and Drug Administration. Towards this goal, high throughput shotgun proteomics experiments (308 cell line/drug combinations +64 control lysates) have been conducted. Using computational network analyses, these proteomic data can be integrated with transcriptomic signatures, generated in tandem, to identify cellular signatures of cardiotoxicity that may predict kinase inhibitor-induced toxicity and enable possible mitigation. Both raw and processed proteomics data have passed several quality control steps and been made publicly available on the PRIDE database. This broad protein kinase inhibitor-stimulated human cardiomyocyte proteomic data and signature set is valuable for prediction of drug toxicities.
Collapse
Affiliation(s)
- Yuguang Xiong
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tong Liu
- Center for Advanced Proteomics Research and Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University - New Jersey Medical School, Newark, NJ, 07103, USA
| | - Tong Chen
- Center for Advanced Proteomics Research and Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University - New Jersey Medical School, Newark, NJ, 07103, USA
| | - Jens Hansen
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bin Hu
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yibang Chen
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gomathi Jayaraman
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Stephan Schürer
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
- Center for Computational Science, University of Miami, Miami, FL, 33136, USA
| | - Dusica Vidovic
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
- Center for Computational Science, University of Miami, Miami, FL, 33136, USA
| | - Joseph Goldfarb
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric A Sobie
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Marc R Birtwistle
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, 29634, USA
| | - Ravi Iyengar
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Hong Li
- Center for Advanced Proteomics Research and Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University - New Jersey Medical School, Newark, NJ, 07103, USA.
| | - Evren U Azeloglu
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|