51
|
Miao C, Cao J, Wang Y, Liu B, Wang Z. Effects of VEGF and VEGFR polymorphisms on the outcome of patients with metastatic renal cell carcinoma treated with sunitinib: a systematic review and meta-analysis. Oncotarget 2017; 8:68854-68862. [PMID: 28978162 PMCID: PMC5620302 DOI: 10.18632/oncotarget.19924] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022] Open
Abstract
To summarize and clarify the association between vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor (VEGFR) polymorphisms and the outcome in patients with metastatic renal cell carcinoma (mRCC) treated with sunitinib. A total of 8 studies including 900 patients were analyzed in this systematic review after screening the database of PubMed, EMBASE and Web of Science. Hazard ratios (HRs) with 95% confidence interval (CI) were used to evaluate the strength of the association. VEGFR1 rs9582036 AA/AC carriers and rs9554320 CC/AC carriers had more favorable overall survival (OS) in patients with mRCC treated with sunitinib (n = 3), but not in progression-free survival (PFS). In addition, VEGFA rs2010963 was associated with poorer PFS of mRCC (n = 1). VEGFA rs699947 was significant in predicting PFS by univariate analysis, but showed no statistical significance in OS (n = 1). VEGFR2 rs1870377 was verified to be associated with sunitinib OS (n = 1). Furthermore, patients with VEGFR3 rs307826 and rs307821 had shorter PFS and OS during sunitinib therapy (n = 2, respectively). Our results suggested that VEGF and VEGFR polymorphisms were associated with outcomes in sunitinib treated mRCC patients, especially VEGFR1 polymorphisms. However, considering the limited study numbers, its clinical application in sunitinib treated mRCC still needs further confirmation.
Collapse
Affiliation(s)
- Chenkui Miao
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingyi Cao
- Department of Urology, Xuzhou Cancer Hospital, Xuzhou, China
| | - Yuhao Wang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bianjiang Liu
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zengjun Wang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
52
|
Diekstra MH, Fritsch A, Kanefendt F, Swen JJ, Moes D, Sörgel F, Kinzig M, Stelzer C, Schindele D, Gauler T, Hauser S, Houtsma D, Roessler M, Moritz B, Mross K, Bergmann L, Oosterwijk E, Kiemeney LA, Guchelaar HJ, Jaehde U. Population Modeling Integrating Pharmacokinetics, Pharmacodynamics, Pharmacogenetics, and Clinical Outcome in Patients With Sunitinib-Treated Cancer. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:604-613. [PMID: 28571114 PMCID: PMC5613186 DOI: 10.1002/psp4.12210] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/01/2017] [Accepted: 05/13/2017] [Indexed: 12/15/2022]
Abstract
The tyrosine kinase inhibitor sunitinib is used as first‐line therapy in patients with metastasized renal cell carcinoma (mRCC), given in fixed‐dose regimens despite its high variability in pharmacokinetics (PKs). Interindividual variability of drug exposure may be responsible for differences in response. Therefore, dosing strategies based on pharmacokinetic/pharmacodynamic (PK/PD) models may be useful to optimize treatment. Plasma concentrations of sunitinib, its active metabolite SU12662, and the soluble vascular endothelial growth factor receptors sVEGFR‐2 and sVEGFR‐3, were measured in 26 patients with mRCC within the EuroTARGET project and 21 patients with metastasized colorectal cancer (mCRC) from the C‐II‐005 study. Based on these observations, PK/PD models with potential influence of genetic predictors were developed and linked to time‐to‐event (TTE) models. Baseline sVEGFR‐2 levels were associated with clinical outcome in patients with mRCC, whereas active drug PKs seemed to be more predictive in patients with mCRC. The models provide the basis of PK/PD‐guided strategies for the individualization of anti‐angiogenic therapies.
Collapse
Affiliation(s)
- M H Diekstra
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - A Fritsch
- Institute of Pharmacy, Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - F Kanefendt
- Institute of Pharmacy, Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - J J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Djar Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - F Sörgel
- IBMP - Institute for Biomedical and Pharmaceutical Research, Nürnberg-Heroldsberg, Germany
| | - M Kinzig
- IBMP - Institute for Biomedical and Pharmaceutical Research, Nürnberg-Heroldsberg, Germany
| | - C Stelzer
- IBMP - Institute for Biomedical and Pharmaceutical Research, Nürnberg-Heroldsberg, Germany
| | - D Schindele
- Department for Urology and Paediatric Urology, University of Magdeburg, Magdeburg, Germany
| | - T Gauler
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - S Hauser
- Department of Urology, University Hospital Bonn, Bonn, Germany
| | - D Houtsma
- Haga Hospital, Den Haag, The Netherlands
| | | | - B Moritz
- CESAR Central Office, Vienna, Austria
| | - K Mross
- Department of Medical Oncology, Tumor Biology Center Freiburg, Freiburg, Germany
| | - L Bergmann
- Cancer-Center Rhein-Main, University Hospital Frankfurt, Frankfurt, Germany
| | - E Oosterwijk
- Department of Urology, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - L A Kiemeney
- Department of Epidemiology and Biostatistics, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - H J Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - U Jaehde
- Institute of Pharmacy, Clinical Pharmacy, University of Bonn, Bonn, Germany
| |
Collapse
|
53
|
Zhou Y, Ingelman-Sundberg M, Lauschke VM. Worldwide Distribution of Cytochrome P450 Alleles: A Meta-analysis of Population-scale Sequencing Projects. Clin Pharmacol Ther 2017; 102:688-700. [PMID: 28378927 PMCID: PMC5600063 DOI: 10.1002/cpt.690] [Citation(s) in RCA: 413] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/28/2017] [Accepted: 03/11/2017] [Indexed: 12/23/2022]
Abstract
Genetic polymorphisms in cytochrome P450 (CYP) genes can result in altered metabolic activity toward a plethora of clinically important medications. Thus, single nucleotide variants and copy number variations in CYP genes are major determinants of drug pharmacokinetics and toxicity and constitute pharmacogenetic biomarkers for drug dosing, efficacy, and safety. Strikingly, the distribution of CYP alleles differs considerably between populations with important implications for personalized drug therapy and healthcare programs. To provide a global distribution map of CYP alleles with clinical importance, we integrated whole‐genome and exome sequencing data from 56,945 unrelated individuals of five major human populations. By combining this dataset with population‐specific linkage information, we derive the frequencies of 176 CYP haplotypes, providing an extensive resource for major genetic determinants of drug metabolism. Furthermore, we aggregated this dataset into spectra of predicted functional variability in the respective populations and discuss the implications for population‐adjusted pharmacological treatment strategies.
Collapse
Affiliation(s)
- Y Zhou
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - M Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - V M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
54
|
Puente J, Laínez N, Dueñas M, Méndez-Vidal MJ, Esteban E, Castellano D, Martinez-Fernández M, Basterretxea L, Juan-Fita MJ, Antón L, León L, Lambea J, Pérez-Valderrama B, Vázquez S, Suarez C, del Muro XG, Gallardo E, Maroto JP, Samaniego ML, Suárez-Paniagua B, Sanz J, Paramio JM, SOGUG (Spanish Oncology Genitourinary Group). Novel potential predictive markers of sunitinib outcomes in long-term responders versus primary refractory patients with metastatic clear-cell renal cell carcinoma. Oncotarget 2017; 8:30410-30421. [PMID: 28423742 PMCID: PMC5444752 DOI: 10.18632/oncotarget.16494] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/24/2017] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Several potential predictive markers of efficacy of targeted agents in patients with metastatic renal cell carcinoma (mRCC) have been identified. Interindividual heterogeneity warrants further investigation. PATIENTS AND METHODS Multicenter, observational, retrospective study in patients with clear-cell mRCC treated with sunitinib. Patients were classified in two groups: long-term responders (LR) (progression-free survival (PFS)≥22 months and at least stable disease), and primary refractory (PR) (progressive disease within 3-months of sunitinib onset). Objectives were to compare baseline clinical factors in both populations and to correlate tumor expression of selected signaling pathways components with sunitinib PFS. RESULTS 123 patients were analyzed (97 LR, 26 PR). In the LR cohort, overall response rate was 79% and median duration of best response was 30 months. Median PFS and overall survival were 43.2 (95% confidence intervals[CI]:37.2-49.3) and 63.5 months (95%CI:55.1-71.9), respectively. At baseline PR patients had a significantly lower proportion of nephrectomies, higher lactate dehydrogenase and platelets levels, lower hemoglobin, shorter time to and higher presence of metastases, and increased Fuhrman grade. Higher levels of HEYL, HEY and HES1 were observed in LR, although only HEYL discriminated populations significantly (AUC[ROC]=0.704; cut-off=34.85). Increased levels of hsa-miR-27b, hsa-miR-23b and hsa-miR-628-5p were also associated with prolonged survival. No statistical significant associations between hsa-miR-23b or hsa-miR-27b and the expression of c-Met were found. CONCLUSIONS Certain mRCC patients treated with sunitinib achieve extremely long-term responses. Favorable baseline hematology values and longer time to metastasis may predict longer PFS. HEYL, hsa-miR-27b, hsa-miR-23b and hsa-miR-628-5p could be potentially used as biomarkers of sunitinib response.
Collapse
Affiliation(s)
- Javier Puente
- Medical Oncology Department, Instituto de Investigación Biomédica, Hospital Clínico Universitario San Carlos, Madrid, Spain
| | - Nuria Laínez
- Medical Oncology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Marta Dueñas
- Molecular Oncology Unit CIEMAT and Instituto Investigación Biomédica, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBERONC, Spain
| | | | - Emilio Esteban
- Medical Oncology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Daniel Castellano
- CIBERONC, Spain
- Medical Oncology Department, and Instituto Investigación Biomédica, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Mónica Martinez-Fernández
- Molecular Oncology Unit CIEMAT and Instituto Investigación Biomédica, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBERONC, Spain
| | | | | | - Luis Antón
- Medical Oncology Department, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Luis León
- Promoción e Planificación da Investigación Sanitaria, Axencia de Coñecemento en Saúde, Santiago de Compostela, Spain
| | - Julio Lambea
- Medical Oncology Department, Hospital Clínico de Zaragoza, Zaragoza, Spain
| | | | - Sergio Vázquez
- Medical Oncology Department, Hospital Universitario Lucus Augusti, Lugo, Spain
| | - Cristina Suarez
- Vall d'Hebron Institute of Oncology, Hospital Universitari Vall d' Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Garcia del Muro
- Medical Oncology Department, Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet, Barcelona, Spain
| | - Enrique Gallardo
- Medical Oncology Department, Hospital Universitari Parc Taulí, Sabadell, Spain
| | - José Pablo Maroto
- Medical Oncology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - M Luz Samaniego
- Statistical Department, Trial Form Support TFS people, Madrid, Spain
| | | | - Julián Sanz
- Pathology Department, Hospital Clínico Universitario San Carlos, Madrid, Spain
| | - Jesús M. Paramio
- Molecular Oncology Unit CIEMAT and Instituto Investigación Biomédica, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBERONC, Spain
| | | |
Collapse
|
55
|
van der Zanden LFM, Vermeulen SH, Oskarsdottir A, Maurits JSF, Diekstra MHM, Ambert V, Cambon-Thomsen A, Castellano D, Fritsch A, Garcia Donas J, Guarch Troyas R, Guchelaar HJ, Hartmann A, Hulsbergen-van de Kaa C, Jaehde U, Junker K, Martinez-Cardus A, Masson G, Oosterwijk-Wakka J, Radu MT, Rafnar T, Rodriguez-Antona C, Roessler M, Ruijtenbeek R, Stefansson K, Warren A, Wessels L, Eisen T, Kiemeney LALM, Oosterwijk E. Description of the EuroTARGET cohort: A European collaborative project on TArgeted therapy in renal cell cancer-GEnetic- and tumor-related biomarkers for response and toxicity. Urol Oncol 2017; 35:529.e9-529.e16. [PMID: 28385611 DOI: 10.1016/j.urolonc.2017.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/04/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE For patients with metastatic renal cell cancer (mRCC), treatment choice is mainly based on clinical parameters. With many treatments available and the limited response to treatment and associated toxicities, there is much interest in identifying better biomarkers for personalized treatment. EuroTARGET aims to identify and characterize host- and tumor-related biomarkers for prediction of response to tyrosine kinase inhibitor therapy in mRCC. Here, we describe the EuroTARGET mRCC patient cohort. METHODS AND MATERIALS EuroTARGET is a European collaborative project designed as an observational study for which patients with mRCC were recruited prospectively in 62 centers. In addition, 462 patients with mRCC from previous studies were included. Detailed clinical information (baseline and follow-up) from all patients was entered in web-based case record forms. Blood was collected for germline DNA and pharmacokinetic/pharmacodynamic analyses and, where available, fresh-frozen tumor material was collected to perform tumor DNA, RNA, kinome, and methylome analyses. RESULTS In total, 1,210 patients with mRCC were included. Of these, 920 received a tyrosine kinase inhibitor as first-line targeted treatment (sunitinib [N = 713, 78%], sorafenib [N = 41, 4%], or pazopanib [N = 166, 18%]) and had at least 6 months of outcome assessment (median follow-up 15.3 months [interquartile range: 8.5-30.2 months]). Germline DNA samples were available from 824 of these patients, fresh-frozen tumor material from 142 patients, fresh-frozen normal kidney tissue from 95 patients, and tissue microarrays created from formalin-fixed paraffin-embedded tumor material from 247 patients. Of the 920 patients, germline DNA variant chip data were successfully generated for 811 patients (Illumina HumanOmniExpress BeadChip). For 80 patients, next-generation exome sequencing of germline and tumor DNA was performed, tumor RNA sequencing was performed for 124 patients, kinome activity measured and processed for 121 patients (PamChip), and methylome data (Illumina Infinium HumanMethylation450 BeadChip) were created for 116 RCC tissues (and 23 normal kidney tissues). For 73 out of the 920 patients, all platform data types were generated. In addition, 40 patients were included in a pharmacokinetic/pharmacodynamic phase IV substudy. CONCLUSIONS Analysis of EuroTARGET cohort data will contribute to personalization of therapy for patients with mRCC. The extensive clinical data and multiplatform EuroTARGET data will be freely available.
Collapse
Affiliation(s)
- Loes F M van der Zanden
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Sita H Vermeulen
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | | | - Jake S F Maurits
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Meta H M Diekstra
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Valentin Ambert
- University of Medicine and Pharmacy Carol Davila, Bucaresti, Romania, Bucuresti, Romania
| | - Anne Cambon-Thomsen
- Epidemiology and analyses in public health, Joint Unit 1027, Institut National de la Santé et de la Recherche Médicale (INSERM), Université Toulouse III Paul Sabatier, Faculty of Medicine, Toulouse, France
| | - Daniel Castellano
- Medical Oncology Department, Hospital Universitario 12 de Octubre, I+12 Research Institute, (CiberOnc), Madrid, Spain
| | - Achim Fritsch
- Institute of Pharmacy, Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - Jesus Garcia Donas
- Medical Oncology, HM Hospitales-Centro Integral Oncológico HM Clara Campal, Madrid, Spain
| | | | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arndt Hartmann
- Department of Pathology, University Erlangen-Nürnberg, Erlangen, Germany
| | - Christina Hulsbergen-van de Kaa
- Department of Pathology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Ulrich Jaehde
- Institute of Pharmacy, Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - Kerstin Junker
- Clinic of Urology and Paediatric Urology, Saarland University, Homburg, Germany
| | - Anna Martinez-Cardus
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Catalonia, Spain
| | | | - Jeannette Oosterwijk-Wakka
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Marius T Radu
- University of Medicine and Pharmacy Carol Davila, Bucaresti, Romania, Bucuresti, Romania
| | | | - Cristina Rodriguez-Antona
- Hereditary Endorine Cancer Group, Spanish National Cancer Research Centre (CNIO) and Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Max Roessler
- CESAR central office, CESAR Central European Society for Anticancer Drug Research-EWIV, Vienna, Austria
| | - Rob Ruijtenbeek
- PamGene International B.V., 's-Hertogenbosch, The Netherlands
| | - Kari Stefansson
- deCODE Genetics/Amgen, Reykjavik, Iceland; Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavík, Iceland
| | - Anne Warren
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Lodewyk Wessels
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Tim Eisen
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Lambertus A L M Kiemeney
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands.
| | - Egbert Oosterwijk
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
56
|
Liu X, Fiocco M, Swen JJ, Guchelaar HJ. Assessment of ethnic differences in sunitinib outcome between Caucasian and Asian patients with metastatic renal cell carcinoma: a meta-analysis. Acta Oncol 2017; 56:582-589. [PMID: 27924664 DOI: 10.1080/0284186x.2016.1265666] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND An increasing number of studies have reported ethnic differences in sunitinib outcome in metastatic renal cell carcinoma (mRCC) patients. However, a comprehensive analysis is still lacking. Therefore, we systematically collected available published data and performed a meta-analysis to compare sunitinib efficacy and toxicity in Asian and Caucasian mRCC patients. METHODS Data were extracted from published results from clinical trials, expanded access program and real-world clinical practice. Progression-free survival (or time to tumor progression), overall survival, objective response rate and adverse events were used as endpoints to evaluate the differences of sunitinib outcome between the two ethnicities. For adverse events, we focused the following clinically relevant side effects: diarrhea, fatigue, mucositis/stomatitis, hand-foot syndrome, hypertension, leukopenia, neutropenia and thrombocytopenia. RESULTS A total of 33 publications including 9977 patients were available for meta-analysis. The efficacy of sunitinib in Asian patients was similar to that in Caucasian patients. However, Asian patients showed a higher incidence of all grades toxicity of hand-foot syndrome, > grade 2 fatigue, > grade 2 hand-foot syndrome and > grade 2 thrombocytopenia. CONCLUSION Ethnic differences in adverse events of sunitinib in mRCC patients existed and dose adjustment in Asian patients may be considered.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, China
| | - Marta Fiocco
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesse J. Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
57
|
Sekar V, Mehrotra DG, Majumder B. Molecular and Functional Diagnostic Tools in Precision Oncology for Urological Malignancies. Indian J Surg Oncol 2017; 8:24-32. [PMID: 28127179 PMCID: PMC5236026 DOI: 10.1007/s13193-016-0591-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/12/2016] [Indexed: 12/29/2022] Open
Abstract
Urological malignancies, represented mainly by prostate, bladder, and renal cancers, are some of the leading causes of cancer-related mortalities worldwide. Despite various efforts over decades to develop early detection tests and effective therapeutic paradigms, the response rate to the existing treatments remains low for both primary and late stage/recurrent phases of these cancers. The evolving landscape of molecular diagnostics, aiming to make the diagnosis and treatment more patient-driven, underpins precision oncology and particularly intends to rationally profile individual tumors and highlight the mechanistic insight and complexity of tumor microenvironment in order to develop biomarkers of toxicity risks and response prediction in a clinically oriented dynamical setting. The present review is an effort to capture some of the recent developments in the area of molecular diagnostics and functional testing platforms and their potential application in clinical decision making in the premises of precision oncology of urological malignancies.
Collapse
Affiliation(s)
- Vasanthakumar Sekar
- Department of Cancer Biology, Mitra Biotech, 202, Narayana Nethralaya, Health City, Hosur Main Road, Bangalore, 560099 India
| | - Debapriya Ghosh Mehrotra
- Department of Molecular Pathology, Mitra Biotech, 202, Narayana Nethralaya, Health City, Hosur Main Road, Bangalore, 560099 India
| | - Biswanath Majumder
- Department of Molecular Pathology, Mitra Biotech, 202, Narayana Nethralaya, Health City, Hosur Main Road, Bangalore, 560099 India
| |
Collapse
|
58
|
Genetic polymorphisms as predictive biomarker of survival in patients with gastrointestinal stromal tumors treated with sunitinib. THE PHARMACOGENOMICS JOURNAL 2017; 18:49-55. [DOI: 10.1038/tpj.2016.83] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 10/17/2016] [Accepted: 10/24/2016] [Indexed: 12/15/2022]
|
59
|
Liu X, Swen JJ, Boven E, Castellano D, Gelderblom H, Mathijssen RH, Rodríguez-Antona C, García-Donas J, Rini BI, Guchelaar HJ. Meta-analysis on the association of VEGFR1 genetic variants with sunitinib outcome in metastatic renal cell carcinoma patients. Oncotarget 2017; 8:1204-1212. [PMID: 27901483 PMCID: PMC5352048 DOI: 10.18632/oncotarget.13597] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 11/07/2016] [Indexed: 11/25/2022] Open
Abstract
VEGFR1 rs9582036 and rs9554320 were previously reported the association with sunitinib progression-free survival (PFS) and overall survival (OS) in patients with metastatic renal cell carcinoma (mRCC). Hereafter, the association of both single nucleotide polymorphisms (SNPs) with PFS/OS was confirmed in two independent mRCC cohorts. The aim of the current study was to validate the associations of both SNPs with sunitinib outcome in three independent well-characterized cohorts (SUTOX, CCF and SOGUG) including 286 sunitinib-treated mRCC patients, as well as to perform a meta-analysis of current and published data combined. We found that rs9582036 and rs9554320 showed a significant association with sunitinib PFS in the CCF cohort (HR: 0.254, 95%CI: 0.092-0.703; P=0.008 and HR: 0.430, 95%CI: 0.200-0.927; P=0.031, respectively). Patients with the variant genotype of rs9582036 and rs9554320 had a shorter median PFS. No significant association of both SNPs with sunitinib PFS or OS was detected in either the SUTOX or SOGUG cohort. After the combination of all available data into a meta-analysis, the association of both SNPs with sunitinib PFS or OS did not achieve the threshold for statistical significance. Our findings suggest that, although VEGFR1 rs9582036 and rs9554320 are involved in sunitinib therapy outcome, its clinical use as biomarkers for prediction of sunitinib outcome in mRCC patients is limited, due to inconsistent findings when analyzing all existing studies together.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, China
| | - Jesse J. Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Epie Boven
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Daniel Castellano
- Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Spanish Oncology Genitourinary Group (SOGUG), Madrid, Spain
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ron H.J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Cristina Rodríguez-Antona
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Jesus García-Donas
- Spanish Oncology Genitourinary Group (SOGUG), Madrid, Spain
- Oncology Unit, Clara Campal Comprehensive Cancer Center, Madrid, Spain
| | - Brian I. Rini
- Department of Solid Tumor Oncology, Cleveland Clinic Taussig Cancer Institute (CCF), Cleveland, Ohio, USA
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
60
|
Numakura K, Tsuchiya N, Kagaya H, Takahashi M, Tsuruta H, Inoue T, Narita S, Huang M, Satoh S, Niioka T, Miura M, Habuchi T. Clinical effects of single nucleotide polymorphisms on drug-related genes in Japanese metastatic renal cell carcinoma patients treated with sunitinib. Anticancer Drugs 2017; 28:97-103. [DOI: 10.1097/cad.0000000000000425] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
61
|
Diekstra MH, Liu X, Swen JJ, Guchelaar HJ. What do we need to make genetic biomarker-guided treatment for renal cell carcinoma a reality? Pharmacogenomics 2017; 18:1-4. [PMID: 27967336 PMCID: PMC5220441 DOI: 10.2217/pgs-2016-0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 10/24/2016] [Indexed: 11/21/2022] Open
Affiliation(s)
- Meta H Diekstra
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Xiaoyan Liu
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
62
|
Perez-Gracia JL, Sanmamed MF, Bosch A, Patiño-Garcia A, Schalper KA, Segura V, Bellmunt J, Tabernero J, Sweeney CJ, Choueiri TK, Martín M, Fusco JP, Rodriguez-Ruiz ME, Calvo A, Prior C, Paz-Ares L, Pio R, Gonzalez-Billalabeitia E, Gonzalez Hernandez A, Páez D, Piulats JM, Gurpide A, Andueza M, de Velasco G, Pazo R, Grande E, Nicolas P, Abad-Santos F, Garcia-Donas J, Castellano D, Pajares MJ, Suarez C, Colomer R, Montuenga LM, Melero I. Strategies to design clinical studies to identify predictive biomarkers in cancer research. Cancer Treat Rev 2016; 53:79-97. [PMID: 28088073 DOI: 10.1016/j.ctrv.2016.12.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 12/19/2016] [Indexed: 12/11/2022]
Abstract
The discovery of reliable biomarkers to predict efficacy and toxicity of anticancer drugs remains one of the key challenges in cancer research. Despite its relevance, no efficient study designs to identify promising candidate biomarkers have been established. This has led to the proliferation of a myriad of exploratory studies using dissimilar strategies, most of which fail to identify any promising targets and are seldom validated. The lack of a proper methodology also determines that many anti-cancer drugs are developed below their potential, due to failure to identify predictive biomarkers. While some drugs will be systematically administered to many patients who will not benefit from them, leading to unnecessary toxicities and costs, others will never reach registration due to our inability to identify the specific patient population in which they are active. Despite these drawbacks, a limited number of outstanding predictive biomarkers have been successfully identified and validated, and have changed the standard practice of oncology. In this manuscript, a multidisciplinary panel reviews how those key biomarkers were identified and, based on those experiences, proposes a methodological framework-the DESIGN guidelines-to standardize the clinical design of biomarker identification studies and to develop future research in this pivotal field.
Collapse
Affiliation(s)
- Jose Luis Perez-Gracia
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IDISNA), Pamplona, Spain.
| | - Miguel F Sanmamed
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ana Bosch
- Division of Oncology and Pathology Department of Clinical Sciences, Lund University, Sweden
| | - Ana Patiño-Garcia
- Department of Pediatrics and CIMA LAB Diagnostics, University Clinic of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Victor Segura
- IDISNA and Bioinformatics Unit, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Navarra, Spain
| | - Joaquim Bellmunt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Josep Tabernero
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Christopher J Sweeney
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Miguel Martín
- Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
| | - Juan Pablo Fusco
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain
| | - Maria Esperanza Rodriguez-Ruiz
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IDISNA), Pamplona, Spain; Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Alfonso Calvo
- Health Research Institute of Navarra (IDISNA), Pamplona, Spain; Department of Histology and Pathology, School of Medicine, University of Navarra, Pamplona, Navarra, Spain
| | - Celia Prior
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ruben Pio
- Health Research Institute of Navarra (IDISNA), Pamplona, Spain; Program in Solid Tumors and Biomarkers, CIMA, University of Navarra, Spain
| | - Enrique Gonzalez-Billalabeitia
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, Universidad Católica San Antonio de Murcia, Murcia, Spain
| | | | - David Páez
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jose María Piulats
- Department of Medical Oncology, Institut Català d'Oncologia, Barcelona, Spain
| | - Alfonso Gurpide
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Mapi Andueza
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain
| | - Guillermo de Velasco
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Roberto Pazo
- Department of Medical Oncology, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Enrique Grande
- Department of Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Pilar Nicolas
- Chair in Law and the Human Genome, University of the Basque Country, Bizkaia, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Service, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, University Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
| | - Jesus Garcia-Donas
- Department of Medical Oncology, HM Hospitales - Centro Integral Oncológico HM Clara Campal, Madrid, Spain
| | - Daniel Castellano
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - María J Pajares
- Health Research Institute of Navarra (IDISNA), Pamplona, Spain; Department of Histology and Pathology, School of Medicine, University of Navarra, Pamplona, Navarra, Spain; Program in Solid Tumors and Biomarkers, CIMA, University of Navarra, Spain
| | - Cristina Suarez
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ramon Colomer
- Department of Oncology, Hospital Universitario de la Princesa, Spain
| | - Luis M Montuenga
- Health Research Institute of Navarra (IDISNA), Pamplona, Spain; Department of Histology and Pathology, School of Medicine, University of Navarra, Pamplona, Navarra, Spain; Program in Solid Tumors and Biomarkers, CIMA, University of Navarra, Spain
| | - Ignacio Melero
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain; Health Research Institute of Navarra (IDISNA), Pamplona, Spain; Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| |
Collapse
|
63
|
Kollmannsberger C. Sunitinib side effects as surrogate biomarkers of efficacy. Can Urol Assoc J 2016; 10:S245-S247. [PMID: 28096937 DOI: 10.5489/cuaj.4315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
With the proliferation of treatment options for the management of metastatic renal cell carcinoma (mRCC) over the past decade, predictive markers of response to therapy are becoming increasingly important. Sunitinib is commonly used in the first-line treatment of mRCC. Common mechanism-based adverse events, including hypertension, hypothyroidism, hand-foot syndrome, and neutropenia, have been explored as potential biomarkers of the clinical efficacy of sunitinib in mRCC and are reviewed in this article.
Collapse
|
64
|
de Velasco G, Gray KP, Hamieh L, Urun Y, Carol HA, Fay AP, Signoretti S, Kwiatkowski DJ, McDermott DF, Freedman M, Pomerantz MM, Choueiri TK. Pharmacogenomic Markers of Targeted Therapy Toxicity in Patients with Metastatic Renal Cell Carcinoma. Eur Urol Focus 2016; 2:633-639. [PMID: 28723497 PMCID: PMC5520643 DOI: 10.1016/j.euf.2016.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/04/2016] [Accepted: 03/26/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND Targeted therapy (TT) in metastatic renal cell carcinoma (mRCC) may be associated with a high rate of toxicity that undermines treatment efficacy and patient quality of life. Polymorphisms in genes involved in the pharmacokinetic pathways of TTs may predict toxicity. OBJECTIVE To investigate whether selected single-nucleotide polymorphisms (SNPs) in three core genes involved in the metabolism and transport of sunitinib and the mTOR inhibitors everolimus and temsirolimus are associated with adverse events (AEs). DESIGN, SETTING, AND PARTICIPANTS Germline DNA was extracted from blood or normal kidney tissue from mRCC patients of Caucasian ethnicity in two cohorts treated with either sunitinib (n=159) or mTOR inhibitors (n=62). Six SNPs in three candidate genes (CYP3A4: rs2242480, rs4646437, and rs2246709; CYP3A5: rs15524; and ABCB1: rs2032582 and rs1045642) were analyzed. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Primary endpoints were grade ≥3 AEs for all patients; grade ≥3 hypertension in the sunitinib cohort, and any grade pneumonitis in the mTOR inhibitors cohort. A logistic regression model was used to assess the association between SNPs and AEs, with adjustment for relevant clinical factors. RESULTS AND LIMITATIONS In total, 221 samples were successfully genotyped for the selected SNPs. In the sunitinib cohort, the CYP3A4 rs464637 AG variant was associated with a lower risk of high-grade AEs (odds ratio 0.27, 95% confidence interval 0.08-0.88; p=0.03), but no SNPs were associated with hypertension. In the mTOR inhibitor cohort, none of the selected SNPs was associated with analyzed toxicities. CONCLUSIONS We observed an association between CYP3A4 polymorphisms and toxicity outcomes in mRCC patients treated with sunitinib, but not with everolimus or temsirolimus. Our findings are exploratory in nature, and further validation in independent and larger cohorts is needed. PATIENT SUMMARY We found that variants of CYP3A4, a gene involved in drug metabolism, are associated with sunitinib toxicity. This information may help in better selection of patients for targeted therapies in metastatic renal cell carcinoma.
Collapse
Affiliation(s)
| | - Kathryn P Gray
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Biostatistics and Computational Biology, Harvard School of Public Health, Boston, MA, USA
| | - Lana Hamieh
- Division of Pulmonary Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Yuksel Urun
- Department of Medical Oncology, Ankara University School of Medicine, Turkey
| | - Hallie A Carol
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andre P Fay
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; PUCRS School of Medicine, Porto Alegre, Brazil
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - David J Kwiatkowski
- Division of Pulmonary Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - David F McDermott
- Department of Medical Oncology, Beth-Israel Deaconess Medical Center, Boston, MA, USA
| | - Matthew Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mark M Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
65
|
Gay C, Toulet D, Le Corre P. Pharmacokinetic drug-drug interactions of tyrosine kinase inhibitors: A focus on cytochrome P450, transporters, and acid suppression therapy. Hematol Oncol 2016; 35:259-280. [DOI: 10.1002/hon.2335] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/04/2016] [Accepted: 07/04/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Caroline Gay
- Pôle Pharmacie; Service Hospitalo-Universitaire de Pharmacie; CHU de Rennes Rennes Cedex France
| | - Delphine Toulet
- Pôle Pharmacie; Service Hospitalo-Universitaire de Pharmacie; CHU de Rennes Rennes Cedex France
| | - Pascal Le Corre
- Pôle Pharmacie; Service Hospitalo-Universitaire de Pharmacie; CHU de Rennes Rennes Cedex France
- Laboratoire de Pharmacie Galénique, Biopharmacie et Pharmacie Clinique; IRSET U1085, Faculté de Pharmacie, Université de Rennes 1; Rennes Cedex France
| |
Collapse
|
66
|
Brown SA, Nhola L, Herrmann J. Cardiovascular Toxicities of Small Molecule Tyrosine Kinase Inhibitors: An Opportunity for Systems-Based Approaches. Clin Pharmacol Ther 2016; 101:65-80. [DOI: 10.1002/cpt.552] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 10/31/2016] [Accepted: 10/31/2016] [Indexed: 12/12/2022]
Affiliation(s)
- S-A Brown
- Department of Cardiovascular Diseases; Mayo Clinic; Rochester Minnesota USA
| | - L Nhola
- Department of Cardiovascular Diseases; Mayo Clinic; Rochester Minnesota USA
| | - J Herrmann
- Department of Cardiovascular Diseases; Mayo Clinic; Rochester Minnesota USA
| |
Collapse
|
67
|
Riazalhosseini Y, Lathrop M. Precision medicine from the renal cancer genome. Nat Rev Nephrol 2016; 12:655-666. [DOI: 10.1038/nrneph.2016.133] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
68
|
Neul C, Schaeffeler E, Sparreboom A, Laufer S, Schwab M, Nies AT. Impact of Membrane Drug Transporters on Resistance to Small-Molecule Tyrosine Kinase Inhibitors. Trends Pharmacol Sci 2016; 37:904-932. [PMID: 27659854 DOI: 10.1016/j.tips.2016.08.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 12/21/2022]
Abstract
Small-molecule inhibitors of tyrosine kinases (TKIs) are the mainstay of treatment for many malignancies and represent novel treatment options for other diseases such as idiopathic pulmonary fibrosis. Twenty-five TKIs are currently FDA-approved and >130 are being evaluated in clinical trials. Increasing evidence suggests that drug exposure of TKIs may significantly contribute to drug resistance, independently from somatic variation of TKI target genes. Membrane transport proteins may limit the amount of TKI reaching the target cells. This review highlights current knowledge on the basic and clinical pharmacology of membrane transporters involved in TKI disposition and their contribution to drug efficacy and adverse drug effects. In addition to non-genetic and epigenetic factors, genetic variants, particularly rare ones, in transporter genes are promising novel factors to explain interindividual variability in the response to TKI therapy.
Collapse
Affiliation(s)
- Claudia Neul
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Germany
| | - Alex Sparreboom
- Division of Pharmaceutics, College of Pharmacy, Ohio State University, Columbus, OH, USA
| | - Stefan Laufer
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Germany; Department of Clinical Pharmacology, Institute of Experimental and Clinical Pharmacology and Toxicology, University Hospital, Tübingen, Germany; Department of Pharmacy and Biochemistry, University of Tübingen, Tübingen, Germany.
| | - Anne T Nies
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, and University of Tübingen, Germany
| |
Collapse
|
69
|
Chae JW, Teo YL, Ho HK, Lee J, Back HM, Yun HY, Karlsson MO, Kwon KI, Chan A. BSA and ABCB1 polymorphism affect the pharmacokinetics of sunitinib and its active metabolite in Asian mRCC patients receiving an attenuated sunitinib dosing regimen. Cancer Chemother Pharmacol 2016; 78:623-32. [PMID: 27485537 DOI: 10.1007/s00280-016-3104-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/06/2016] [Indexed: 11/25/2022]
Abstract
PURPOSE An attenuated dosing (AD) sunitinib regimen of 37.5 mg daily has been suggested to reduce the toxicity reported with the standard dosing regimen to metastatic renal cell carcinoma (mRCC) patients. The aim of this study was to characterize the population pharmacokinetic (PK) properties of sunitinib and SU12662, the active metabolite, in patients receiving the AD regimen and to ascertain significant covariates influencing PK parameters. METHODS Thirty-one mRCC patients receiving AD sunitinib regimen were included. Plasma samples were collected on day 29 of each treatment cycle after the start of the therapy. Nonlinear mixed-effects modeling was applied to estimate the population PK properties of sunitinib and SU12662 as well as the effect of covariates on PK parameters. Monte Carlo simulation was also performed to predict the total trough level (TTL) of sunitinib and SU12662. RESULTS Sunitinib population means for CL/F and V d /F central were 13.8 L/h and 1720 L, respectively. SU12662 population means for CL/F and V d /F were 42.1 L/h and 1410 L, respectively. Body surface area (BSA) and ABCB1 polymorphism significantly influenced the CL/F variability of sunitinib: CL/F parent = 13.8 × exp((BSA - 1.75) × 2.08 + (ABCB1 genotype - 0.67) × 0.61), ABCB1-0: wild genotype, 1: mutant genotype. The effect size of ABCB1 mutant genotype and BSA greater than 1.75 m(2) in relation to sunitinib clearance was 31.14 % (p = 0.006) and 22.11 % (p = 0.011), respectively, relative to the reference group. CONCLUSIONS Adjusting doses of sunitinib according to BSA and ABCB1 polymorphism in Asian mRCC patients may be recommended for sufficient attainment of a target TTL of sunitinib and its metabolite.
Collapse
Affiliation(s)
- Jung-Woo Chae
- College of Pharmacy, Chungnam National University, Daejeon, 305-764, Korea.,Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Block S4A, Singapore, 117543, Singapore
| | - Yi Ling Teo
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Block S4A, Singapore, 117543, Singapore
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Block S4A, Singapore, 117543, Singapore
| | - Jaeyeon Lee
- College of Pharmacy, Chungnam National University, Daejeon, 305-764, Korea
| | - Hyun-Moon Back
- College of Pharmacy, Chungnam National University, Daejeon, 305-764, Korea
| | - Hwi-Yeol Yun
- College of Pharmacy, Chungnam National University, Daejeon, 305-764, Korea
| | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Kwang-Il Kwon
- College of Pharmacy, Chungnam National University, Daejeon, 305-764, Korea.
| | - Alexandre Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Block S4A, Singapore, 117543, Singapore. .,Oncology Pharmacy, National Cancer Centre Singapore, Singapore, 169610, Singapore.
| |
Collapse
|
70
|
García-Donas J, Beuselinck B, Inglada-Pérez L, Graña O, Schöffski P, Wozniak A, Bechter O, Apellániz-Ruiz M, Leandro-García LJ, Esteban E, Castellano DE, González Del Alba A, Climent MA, Hernando S, Arranz JA, Morente M, Pisano DG, Robledo M, Rodriguez-Antona C. Deep sequencing reveals microRNAs predictive of antiangiogenic drug response. JCI Insight 2016; 1:e86051. [PMID: 27699216 DOI: 10.1172/jci.insight.86051] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The majority of metastatic renal cell carcinoma (RCC) patients are treated with tyrosine kinase inhibitors (TKI) in first-line treatment; however, a fraction are refractory to these antiangiogenic drugs. MicroRNAs (miRNAs) are regulatory molecules proven to be accurate biomarkers in cancer. Here, we identified miRNAs predictive of progressive disease under TKI treatment through deep sequencing of 74 metastatic clear cell RCC cases uniformly treated with these drugs. Twenty-nine miRNAs were differentially expressed in the tumors of patients who progressed under TKI therapy (P values from 6 × 10-9 to 3 × 10-3). Among 6 miRNAs selected for validation in an independent series, the most relevant associations corresponded to miR-1307-3p, miR-155-5p, and miR-221-3p (P = 4.6 × 10-3, 6.5 × 10-3, and 3.4 × 10-2, respectively). Furthermore, a 2 miRNA-based classifier discriminated individuals with progressive disease upon TKI treatment (AUC = 0.75, 95% CI, 0.64-0.85; P = 1.3 × 10-4) with better predictive value than clinicopathological risk factors commonly used. We also identified miRNAs significantly associated with progression-free survival and overall survival (P = 6.8 × 10-8 and 7.8 × 10-7 for top hits, respectively), and 7 overlapped with early progressive disease. In conclusion, this is the first miRNome comprehensive study, to our knowledge, that demonstrates a predictive value of miRNAs for TKI response and provides a new set of relevant markers that can help rationalize metastatic RCC treatment.
Collapse
Affiliation(s)
- Jesús García-Donas
- Oncology Unit, HM Hospitales - Centro Integral Oncológico HM Clara Campal, Madrid, Spain.,Spanish Oncology Genitourinary Group, Madrid, Spain
| | - Benoit Beuselinck
- Department of General Medical Oncology, University Hospitals Leuven, and.,Laboratory for Experimental Oncology, KU Leuven, Leuven, Belgium
| | - Lucía Inglada-Pérez
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| | - Osvaldo Graña
- Bioinformatics Unit, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Patrick Schöffski
- Department of General Medical Oncology, University Hospitals Leuven, and.,Laboratory for Experimental Oncology, KU Leuven, Leuven, Belgium
| | | | - Oliver Bechter
- Department of General Medical Oncology, University Hospitals Leuven, and.,Laboratory for Experimental Oncology, KU Leuven, Leuven, Belgium
| | - Maria Apellániz-Ruiz
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Luis Javier Leandro-García
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Emilio Esteban
- Spanish Oncology Genitourinary Group, Madrid, Spain.,Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Daniel E Castellano
- Spanish Oncology Genitourinary Group, Madrid, Spain.,Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Aranzazu González Del Alba
- Spanish Oncology Genitourinary Group, Madrid, Spain.,Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Miguel Angel Climent
- Spanish Oncology Genitourinary Group, Madrid, Spain.,Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Susana Hernando
- Spanish Oncology Genitourinary Group, Madrid, Spain.,Hospital Universitario Fundación Alcorcón, Madrid, Spain
| | - José Angel Arranz
- Spanish Oncology Genitourinary Group, Madrid, Spain.,Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Manuel Morente
- Tumour Bank Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - David G Pisano
- Bioinformatics Unit, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| | - Cristina Rodriguez-Antona
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| |
Collapse
|
71
|
Dornbusch J, Walter M, Gottschalk A, Obaje A, Junker K, Ohlmann CH, Meinhardt M, Zacharis A, Zastrow S, Schoffer O, Grimm MO, Klug SJ, Wirth MP, Fuessel S. Evaluation of polymorphisms in angiogenesis-related genes as predictive and prognostic markers for sunitinib-treated metastatic renal cell carcinoma patients. J Cancer Res Clin Oncol 2016; 142:1171-82. [PMID: 26935927 DOI: 10.1007/s00432-016-2137-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/18/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE Single nucleotide polymorphisms (SNPs) in angiogenesis-associated genes might play an important role in activity of the tyrosine kinase inhibitor sunitinib and could affect survival of cancer patients treated with this drug. The aim of this retrospective study was to elucidate the role of 10 known SNPs in VEGFA, VEGFR1, VEGFR2 and VEGFR3 as potential prognostic and predictive markers in an independent cohort of patients with metastatic renal cell carcinoma (mRCC). METHODS DNA from 121 mRCC patients treated with sunitinib was used to analyze SNPs by TaqMan genotyping assays. Disease control rate was evaluated according to RECIST. Adverse effects of sunitinib were registered from medical records. The results of Cox and logistic regression were verified by correction for multiple testing. RESULTS Kaplan-Meier analysis revealed a reduced progression-free survival in patients with the wild-type (WT) allele of the VEGFA SNP rs699947 compared to variant alleles. Patients with the AA/AC-alleles of the VEGFR1 SNP rs9582036 had an improved median overall survival compared to those with the CC-WT allele what could be confirmed by multivariable Cox proportional hazard regression analyses. No statistically significant associations between the analyzed SNPs and higher risk for adverse effects were observed. CONCLUSIONS The results of this study suggest that most of the selected SNPs in angiogenesis-related genes are not associated with survival of mRCC patients after sunitinib therapy or with adverse effects. Only the VEGFR1 SNP rs9582036 showed a statistically significant association with overall survival. The potential of SNPs as prognostic and predictive markers for sunitinib-treated mRCC patients should be finally assessed by prospective studies.
Collapse
Affiliation(s)
- Juana Dornbusch
- Department of Urology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Martina Walter
- Department of Urology, University Hospital of Jena, Lessingstr. 1, 07743, Jena, Germany
| | - Andrea Gottschalk
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Alice Obaje
- Department of Urology, University Hospital of Jena, Lessingstr. 1, 07743, Jena, Germany
| | - Kerstin Junker
- Department of Urology, Saarland University Medical Center, Kirrberger Str. 1, 66424, Homburg, Germany
| | - Carsten-Henning Ohlmann
- Department of Urology, Saarland University Medical Center, Kirrberger Str. 1, 66424, Homburg, Germany
| | - Matthias Meinhardt
- Institute of Pathology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Aristeidis Zacharis
- Department of Urology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Stefan Zastrow
- Department of Urology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Olaf Schoffer
- Cancer Epidemiology, University Cancer Center Dresden, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Marc-Oliver Grimm
- Department of Urology, University Hospital of Jena, Lessingstr. 1, 07743, Jena, Germany
| | - Stefanie J Klug
- Cancer Epidemiology, University Cancer Center Dresden, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Manfred P Wirth
- Department of Urology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Susanne Fuessel
- Department of Urology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| |
Collapse
|
72
|
Bianconi M, Faloppi L, Lopez-Beltran A, Scarpelli M, Scartozzi M, Cheng L, Montironi R. Clinical impact of tumoral angiogenesis on renal cell carcinoma management: where do we stand? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1181974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
73
|
Heng DYC, Choueiri TK. The evolving landscape of metastatic renal cell carcinoma. Am Soc Clin Oncol Educ Book 2016:299-302. [PMID: 24451753 DOI: 10.14694/edbook_am.2012.32.25] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The treatment paradigm in metastatic renal cell carcinoma (mRCC) has evolved over the last 5 years. There are now seven approved targeted therapies against the vascular endothelial growth factor (VEGF) and mammalian target of rapamycin (mTOR) pathways. The use of targeted therapy, sequences, combinations, and investigational compounds will be discussed. Prognostic and predictive tools are detailed, although much work must be done to find predictive biomarkers in an effort to individualize therapy for patients.
Collapse
Affiliation(s)
- Daniel Y C Heng
- From the Tom Baker Cancer Center, University of Calgary, Calgary, Alberta, Canada; Dana Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Toni K Choueiri
- From the Tom Baker Cancer Center, University of Calgary, Calgary, Alberta, Canada; Dana Farber Cancer Institute and Harvard Medical School, Boston, MA
| |
Collapse
|
74
|
Nelson MR, Johnson T, Warren L, Hughes AR, Chissoe SL, Xu CF, Waterworth DM. The genetics of drug efficacy: opportunities and challenges. Nat Rev Genet 2016; 17:197-206. [PMID: 26972588 DOI: 10.1038/nrg.2016.12] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Lack of sufficient efficacy is the most common cause of attrition in late-phase drug development. It has long been envisioned that genetics could drive stratified drug development by identifying those patient subgroups that are most likely to respond. However, this vision has not been realized as only a small proportion of drugs have been found to have germline genetic predictors of efficacy with clinically meaningful effects, and so far all but one were found after drug approval. With the exception of oncology, systematic application of efficacy pharmacogenetics has not been integrated into drug discovery and development across the industry. Here, we argue for routine, early and cumulative screening for genetic predictors of efficacy, as an integrated component of clinical trial analysis. Such a strategy would identify clinically relevant predictors that may exist at the earliest possible opportunity, allow these predictors to be integrated into subsequent clinical development and provide mechanistic insights into drug disposition and patient-specific factors that influence response, therefore paving the way towards more personalized medicine.
Collapse
Affiliation(s)
- Matthew R Nelson
- Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania 19406, USA
| | - Toby Johnson
- Target Sciences, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Liling Warren
- GlaxoSmithKline, Durham, North Carolina 27713, USA
- Acclarogen, Cambridge CB4 0WS, UK
| | - Arlene R Hughes
- PAREXEL International, Research Triangle Park, North Carolina 27713, USA
| | | | - Chun-Fang Xu
- Target Sciences, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Dawn M Waterworth
- Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania 19406, USA
| |
Collapse
|
75
|
Minardi D, Quaresima L, Santoni M, Bianconi M, Scartozzi M, Cascinu S, Muzzonigro G. Recent aspects of sunitinib therapy in patients with metastatic clear-cell renal cell carcinoma: a systematic review of the literature. Curr Urol Rep 2016; 16:3. [PMID: 25627021 DOI: 10.1007/s11934-014-0478-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sunitinib is an orally available inhibitor of multiple tyrosine-kinase receptors approved for the treatment of advanced clear-cell renal cell carcinoma (ccRCC), a disease which has habitually had a very poor patient survival rate. Although it has become the most widely used drug for this disease, it remains not completely clear the best treatment strategy with these agent. The aim of this review is to highlight the most recent and interesting aspects of the research on treatment of advanced ccRCC with sunitinib and eventually determine alternative treatment schedule to reduce the incidence of side effects; we also wanted to review recent biomarkers able to predict response to therapy and also to point out the mechanism of acquired resistance to this drug.
Collapse
Affiliation(s)
- Daniele Minardi
- Department of Clinic and Specialistic Sciences - Urology, Polytechnic University of the Marche Region - Azienda Ospedaliero - Universitaria Ospedali Riuniti, via Conca, 71, 60131, Ancona, Italy,
| | | | | | | | | | | | | |
Collapse
|
76
|
Wolking S, Schaeffeler E, Lerche H, Schwab M, Nies AT. Impact of Genetic Polymorphisms of ABCB1 (MDR1, P-Glycoprotein) on Drug Disposition and Potential Clinical Implications: Update of the Literature. Clin Pharmacokinet 2016; 54:709-35. [PMID: 25860377 DOI: 10.1007/s40262-015-0267-1] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
ATP-binding cassette transporter B1 (ABCB1; P-glycoprotein; multidrug resistance protein 1) is an adenosine triphosphate (ATP)-dependent efflux transporter located in the plasma membrane of many different cell types. Numerous structurally unrelated compounds, including drugs and environmental toxins, have been identified as substrates. ABCB1 limits the absorption of xenobiotics from the gut lumen, protects sensitive tissues (e.g. the brain, fetus and testes) from xenobiotics and is involved in biliary and renal secretion of its substrates. In recent years, a large number of polymorphisms of the ABCB1 [ATP-binding cassette, sub-family B (MDR/TAP), member 1] gene have been described. The variants 1236C>T (rs1128503, p.G412G), 2677G>T/A (rs2032582, p.A893S/T) and 3435C>T (rs1045642, p.I1145I) occur at high allele frequencies and create a common haplotype; therefore, they have been most widely studied. This review provides an overview of clinical studies published between 2002 and March 2015. In summary, the effect of ABCB1 variation on P-glycoprotein expression (messenger RNA and protein expression) and/or activity in various tissues (e.g. the liver, gut and heart) appears to be small. Although polymorphisms and haplotypes of ABCB1 have been associated with alterations in drug disposition and drug response, including adverse events with various ABCB1 substrates in different ethnic populations, the results have been majorly conflicting, with limited clinical relevance. Future research activities are warranted, considering a deep-sequencing approach, as well as well-designed clinical studies with appropriate sample sizes to elucidate the impact of rare ABCB1 variants and their potential consequences for effect sizes.
Collapse
Affiliation(s)
- Stefan Wolking
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Hoppe-Seyler Strasse 3, 72076, Tübingen, Germany
| | | | | | | | | |
Collapse
|
77
|
Diekstra MHM, Swen JJ, Gelderblom H, Guchelaar HJ. A decade of pharmacogenomics research on tyrosine kinase inhibitors in metastatic renal cell cancer: a systematic review. Expert Rev Mol Diagn 2016; 16:605-18. [PMID: 26837796 DOI: 10.1586/14737159.2016.1148601] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The individual response to targeted tyrosine kinase inhibitors (TKIs) in the treatment of metastatic renal cell cancer (mRCC) is highly variable. Outlined in this article are findings on potential biomarkers for TKI treatment outcome in mRCC and an evaluation of the status of clinical implementation. METHODS Articles were selected by two independent reviewers using a systematic search in five medical databases on renal cell carcinoma, TKIs, and pharmacogenetics. RESULTS Many researchers have focused on predictive biomarkers for treatment outcome of targeted therapies in mRCC patients. Attempts to explain differences in efficacy and toxicity of TKIs by use of genetic variants in genes related to the pharmacokinetics and pharmacodynamics of the drug have been successful. CONCLUSION Most findings on potential biomarkers have not been validated and therefore biomarker testing to guide choice of therapy and dose in mRCC is not yet feasible.
Collapse
Affiliation(s)
- Meta H M Diekstra
- a Department of Clinical Pharmacy and Toxicology , Leiden University Medical Center , Leiden , Netherlands
| | - Jesse J Swen
- a Department of Clinical Pharmacy and Toxicology , Leiden University Medical Center , Leiden , Netherlands
| | - Hans Gelderblom
- b Department of Medical Oncology , Leiden University Medical Center , Leiden , Netherlands
| | - Henk-Jan Guchelaar
- a Department of Clinical Pharmacy and Toxicology , Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
78
|
Qin C, Cao Q, Li P, Wang S, Wang J, Wang M, Chu H, Zhou L, Li X, Ye D, Zhang H, Huang Y, Dong B, Sun X, Zou Q, Cai H, Sun L, Zhu J, Liu F, Ji J, Cui L, Wang X, Zhou H, Zhao H, Wu B, Chen J, Jiang M, Zhang Z, Shao P, Ju X, Yin C. The influence of genetic variants of sorafenib on clinical outcomes and toxic effects in patients with advanced renal cell carcinoma. Sci Rep 2016; 6:20089. [PMID: 26830973 PMCID: PMC4735712 DOI: 10.1038/srep20089] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 12/07/2015] [Indexed: 01/26/2023] Open
Abstract
The purpose of the present study was to investigate whether genetic variants that influence angiogenesis and sorafenib pharmacokinetics are associated with clinical outcomes and toxic effects in advanced renal cell carcinoma patients treated with this drug. One hundred patients with advanced renal cell carcinoma were enrolled. Forty-two polymorphisms in 15 genes were selected for genotyping and analyzed for associations with progression-free survival, overall survival, and toxic effects. We found that rs1570360 in VEGF and rs2239702 in VEGFR2 were significantly associated with progression-free. Specifically, patients carrying the variant genotypes (AG + AA) of these two polymorphisms both had an unfavorable progression-free. In addition, compared with those with the rs2239702 GG genotype, patients with the AG + AA genotype suffered an unfavorable OS. We found that the VEGF rs2010963 CG + GG genotypes had a significantly increased risk of hand-foot syndrome, and the ABCB1 rs1045642 CT + TT genotypes had an increased risk of high blood pressure. Our results suggest that polymorphisms in VEGF and VEGFR2 are associated with sorafenib clinical outcomes, and polymorphisms in VEGF and ABCB1 are associated with sorafenib-related toxicities. Larger studies are warranted to validate our findings.
Collapse
Affiliation(s)
- Chao Qin
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiang Cao
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pu Li
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shangqian Wang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Wang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meilin Wang
- Cancer Center of Nanjing Medical University, Department of Molecular &Genetic Toxicology, Nanjing Medical University, Nanjing, China
| | - Haiyan Chu
- Cancer Center of Nanjing Medical University, Department of Molecular &Genetic Toxicology, Nanjing Medical University, Nanjing, China
| | - Liqun Zhou
- Department of Urology, Peking University Fist Hospital, Beijing, China
| | - Xuesong Li
- Department of Urology, Peking University Fist Hospital, Beijing, China
| | - Dingwei Ye
- Department of Urology, Fu Dan University Shanghai Cancer Center, Shanghai, China
| | - Hailiang Zhang
- Department of Urology, Fu Dan University Shanghai Cancer Center, Shanghai, China
| | - Yiran Huang
- Department of Urology, Shanghai Renji Hospital, Shanghai, China
| | - Baijun Dong
- Department of Urology, Shanghai Renji Hospital, Shanghai, China
| | - Xiaofeng Sun
- Department ofOncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Qing Zou
- Department of Urology, Jiangsu Cancer Hospital, Nanjing, China
| | - Hongzhou Cai
- Department of Urology, Jiangsu Cancer Hospital, Nanjing, China
| | - Lijiang Sun
- Department of Urology, the Affiliated Hospital of Qing Dao University, Qiang Dao, China
| | - Jian Zhu
- Department of Urology, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Fade Liu
- Department of Urology, Nanjing Jiangning Hospital, Nanjing, China
| | - Junbiao Ji
- Department of Urology, Nanjing Jiangning Hospital, Nanjing, China
| | - Li Cui
- Department of Urology, the First People's Hospital of Changzhou, Changzhou, China
| | - Xiaoxiang Wang
- Department of Urology, Yangzhou NO.1 People's Hospital, Yangzhou, China
| | - Hai Zhou
- Department of Urology, Yangzhou NO.1 People's Hospital, Yangzhou, China
| | - Hu Zhao
- Department of Urology, Jiangyin People's Hospital, Jiangyin, China
| | - Bin Wu
- Department of Urology, Jiangyin People's Hospital, Jiangyin, China
| | - Jianchun Chen
- Department of Urology, Wujiang NO.1 People's Hospital, Wujiang, China
| | - Minjun Jiang
- Department of Urology, Wujiang NO.1 People's Hospital, Wujiang, China
| | - Zhengdong Zhang
- Cancer Center of Nanjing Medical University, Department of Molecular &Genetic Toxicology, Nanjing Medical University, Nanjing, China
| | - Pengfei Shao
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaobing Ju
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Changjun Yin
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
79
|
Sunitinib-induced hypertension in CYP3A4 rs4646437 A-allele carriers with metastatic renal cell carcinoma. THE PHARMACOGENOMICS JOURNAL 2016; 17:42-46. [PMID: 26810136 DOI: 10.1038/tpj.2015.100] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 08/27/2015] [Accepted: 10/16/2015] [Indexed: 11/08/2022]
Abstract
The single nucleotide polymorphism (SNP) rs4646437G>A in CYP3A4 was suggested to be related to sunitinib toxicity. Our objective was to perform an in-depth investigation of the association between this SNP and sunitinib toxicity and efficacy using a large cohort of metastatic renal cell carcinoma (mRCC) patients. We collected DNA and clinical information of mRCC patients treated with sunitinib. SNP rs4646437 in CYP3A4 was tested for associations with toxicity using logistic regression. Cox regression modeling was used for association analysis of rs4646437 with progression-free survival (PFS) and overall survival (OS). In a total of 287 patients, the A-allele of CYP3A4 rs4646437 was associated with an increased risk for hypertension (odds ratio=2.4, 95% confidence interval: 1.1-5.2, P=0.021) and showed no significant association with PFS or OS. In conclusion, hypertension is more likely to occur in A-allele carriers of the CYP3A4 rs4646437 variant in our cohort of mRCC patients treated with sunitinib.
Collapse
|
80
|
English PA, Williams JA, Martini JF, Motzer RJ, Valota O, Buller RE. A case for the use of receiver operating characteristic analysis of potential clinical efficacy biomarkers in advanced renal cell carcinoma. Future Oncol 2015; 12:175-82. [PMID: 26674983 PMCID: PMC5549778 DOI: 10.2217/fon.15.290] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
AIM Assess patient-level utility of suggested pretreatment biomarkers of sunitinib in advanced renal cell carcinoma. PATIENTS & METHODS Kaplan-Meier analysis of data from a randomized, Phase II study (n = 292) suggested baseline predictive value for circulating soluble Ang-2 and MMP-2 and HIF-1α percentage of tumor expression. Using this dataset, the sensitivity, specificity and area under the curve (AUC) were calculated, using receiver operating characteristic (ROC) curves. RESULTS Based on a ROC (sensitivity vs 1 - specificity) threshold AUC value of >0.8, neither Ang-2 (0.67) nor MMP-2 (0.65), nor HIF-1α percentage of tumor expression (0.65), performed appropriately from a patient-selection standpoint. CONCLUSION To properly assess potential biomarkers, sensitivity and specificity characteristics should be obtained by ROC analysis.
Collapse
Affiliation(s)
| | - J Andrew Williams
- Pfizer Oncology, 10646 Science Center Drive, San Diego, CA 92121, USA
| | | | - Robert J Motzer
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | - Olga Valota
- Pfizer Oncology, via AM Mozzoni 12, Milan, 20152, Italy
| | - Richard E Buller
- Pfizer Oncology, 10646 Science Center Drive, San Diego, CA 92121, USA
| |
Collapse
|
81
|
Edesa WA, Abdelmalek RR. Efficacy and toxicity of sunitinib in metastatic renal cell carcinoma patients in Egypt. Asian Pac J Cancer Prev 2015; 16:1971-6. [PMID: 25773796 DOI: 10.7314/apjcp.2015.16.5.1971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To evaluate our results in terms of response, survival and toxicity profile of sunitinib among Egyptian patients with metastatic renal cell carcinoma. MATERIALS AND METHODS Between January 2010 and December 2013, 44 patients with metastatic renal cell carcinoma who received sunitinib at an oncology center of Cairo university hospitals were enrolled in this retrospective analysis. RESULTS The median age of the patients was 53 years, 22 (50%) having localized disease at presentation ,while the remaining half of the patients presented with metastasis. At a median follow up of 19 months, 9 (21%) patients achieved partial remission, while disease was reported stable in 20 cases (45%) and progressive in 7 (16%), 4 (9%) being lost to follow up, and 4 (9%) had discontinued therapy due to toxicity. The median overall survival was 23 months (95%CI 15.2 - 30.9), while progression free survival was 12 months (95%CI 11.6 - 12.3). The most commonly reported non hematological grade 3 adverse events included mucositis (15.9%), hand-foot syndrome (13.6%), and fatigue (9%), while the predominant grade 3 or 4 laboratory abnormalities were neutropenia (6.8%), followed by anemia in 4.5% of patients. CONCLUSIONS Our efficacy data were comparable to the published literature in terms of progression free survival and overall survival , while toxicity profile is different from Asian and western countries. However, sunitinib adverse events were manageable and tolerable in most of our Egyptian patients.
Collapse
Affiliation(s)
- Wael Abdelgawad Edesa
- Kasr Alaini Center of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt E-mail :
| | | |
Collapse
|
82
|
Donskov F, Michaelson MD, Puzanov I, Davis MP, Bjarnason GA, Motzer RJ, Goldstein D, Lin X, Cohen DP, Wiltshire R, Rini BI. Sunitinib-associated hypertension and neutropenia as efficacy biomarkers in metastatic renal cell carcinoma patients. Br J Cancer 2015; 113:1571-80. [PMID: 26492223 PMCID: PMC4705883 DOI: 10.1038/bjc.2015.368] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 08/18/2015] [Accepted: 09/23/2015] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Metastatic renal cell carcinoma (mRCC) prognostic models may be improved by incorporating treatment-induced toxicities. METHODS In sunitinib-treated mRCC patients (N=770), baseline prognostic factors and treatment-induced toxicities (hypertension (systolic blood pressure ⩾140 mm Hg), neutropenia (grade ⩾2), thrombocytopenia (grade ⩾2), hand-foot syndrome (grade >0), and asthenia/fatigue (grade >0)) were analysed in multivariate analyses of progression-free survival (PFS) and overall survival (OS) end points. RESULTS On-treatment neutropenia and hypertension were associated with longer PFS (P=0.0276 and P<0.0001, respectively) and OS (P=0.0014 and P<0.0001, respectively), independent of baseline prognostic factors, including International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) criteria. By 12-week landmark analysis, neutropenia was significantly associated with longer PFS and OS (P=0.013 and P=0.0122, respectively) and hypertension or hand-foot syndrome with longer OS (P=0.0036 and P=0.0218, respectively). The concordance index was 0.65 (95% CI: 0.63-0.67) for IMDC classification alone and 0.72 (95% CI: 0.70-0.74) when combined with hypertension and neutropenia. Considering hypertension and neutropenia (developing both vs neither) changed IMDC-predicted median OS in each IMDC risk group (favourable: 45.3 vs 19.5 months; intermediate: 32.5 vs 8.0 months; poor: 21.1 vs 4.8 months). CONCLUSIONS On-treatment neutropenia and hypertension are independent biomarkers of sunitinib efficacy and may add prognostic accuracy to the IMDC model.
Collapse
Affiliation(s)
- Frede Donskov
- Department of Oncology, Aarhus University Hospital, Norrebrogade 44, Aarhus C 8000, Denmark
| | - M Dror Michaelson
- Massachusetts General Hospital Cancer Center, The Claire and John Bertucci Center for Genitourinary Cancers, 55 Fruit Street, Yawkey 7E, Boston, MA 02114, USA
| | - Igor Puzanov
- Division of Hematology–Oncology, Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN 37232-6307, USA
| | - Mellar P Davis
- Palliative Medicine and Supportive Oncology Services, Division of Solid Tumor, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, Desk R35, Cleveland, OH 44195, USA
| | - Georg A Bjarnason
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada
| | - Robert J Motzer
- Department of Medicine, Genitourinary Oncology Service, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | - David Goldstein
- Department of Medical Oncology, Prince of Wales Hospital, Randwick, NSW 2031, Australia
| | - Xun Lin
- Pfizer Oncology, 10646 Science Center Drive, La Jolla, CA 92121, USA
| | - Darrel P Cohen
- Pfizer Oncology, 10646 Science Center Drive, La Jolla, CA 92121, USA
| | - Robin Wiltshire
- Pfizer Oncology, Walton Oaks, Dorking Road, Walton-on-the-Hill, Tadworth, Surrey KT20 7NS, UK
| | - Brian I Rini
- Cleveland Clinic Taussig Cancer Institute, Glickman Urological Institute, 9500 Euclid Avenue, Desk R35, Cleveland, OH 44195, USA
| |
Collapse
|
83
|
Winer AG, Motzer RJ, Hakimi AA. Prognostic Biomarkers for Response to Vascular Endothelial Growth Factor-Targeted Therapy for Renal Cell Carcinoma. Urol Clin North Am 2015; 43:95-104. [PMID: 26614032 DOI: 10.1016/j.ucl.2015.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Improved understanding of renal carcinoma disease biology has led to the discovery and approval of five novel therapies targeting specific molecules in the vascular endothelial growth factor (VEGF) biochemical pathway. Biomarker studies attempting to predict response to VEGF-targeted therapies have largely focused on circulating proteins, tissue-based molecules, and germline polymorphisms. Thus far studies have yielded conflicting results that require prospective validation; therefore no definitive biomarker has yet been integrated into the clinician's armamentarium. However, early analyses featuring genomic biomarkers have generated promising findings. This article provides an overview of available biomarkers evaluated with respect to VEGF-targeted therapies in patients with advanced renal cell carcinoma.
Collapse
Affiliation(s)
- Andrew G Winer
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 353 East 68th Street, New York, NY 10065, USA
| | - Robert J Motzer
- Genitourinary Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - A Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 353 East 68th Street, New York, NY 10065, USA.
| |
Collapse
|
84
|
Mickley A, Kovaleva O, Kzhyshkowska J, Gratchev A. Molecular and immunologic markers of kidney cancer-potential applications in predictive, preventive and personalized medicine. EPMA J 2015; 6:20. [PMID: 26500709 PMCID: PMC4617448 DOI: 10.1186/s13167-015-0042-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/18/2015] [Indexed: 12/12/2022]
Abstract
Kidney cancer is one of the deadliest malignancies due to frequent late diagnosis (33 % or renal cell carcinoma are metastatic at diagnosis) and poor treatment options. There are two major subtypes of kidney cancer: renal cell carcinoma (RCC) and renal pelvis carcinoma. The risk factors for RCC, accounting for more than 90 % of all kidney cancers, are smoking, obesity, hypertension, misuse of pain medication, and some genetic diseases. The most common molecular markers of kidney cancer include mutations and epigenetic inactivation of von Hippel-Lindau (VHL) gene, genes of vascular endothelial growth factor (VEGF) pathway, and carbonic anhydrase IX (CIAX). The role of epigenetic pathways, including DNA methylation and chromatin structure remodeling, was also demonstrated. Immunologic properties of RCC enable this type of tumor to escape immune response effectively. An important role in this process is played by tumor-associated macrophages that demonstrate mixed M1/M2 phenotype. In this review, we discuss molecular and cellular aspects for RCC development and current state of knowledge allowing personalized approaches for diagnostics and prognostic prediction of this disease. A set of macrophage markers is suggested for the analysis of the association of macrophage phenotype and disease prognosis.
Collapse
Affiliation(s)
- Amanda Mickley
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | | | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany ; Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany ; Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
| | - Alexei Gratchev
- Blokhin Cancer Research Center, Moscow, Russia ; Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia ; Laboratory of the Tumour Stromal Cells Biology, Institute of Carcinogenesis, Blokhin Cancer Research Center, Kashirskoye Shosse 24, Moscow, Russia
| |
Collapse
|
85
|
Diekstra MH, Swen JJ, Boven E, Castellano D, Gelderblom H, Mathijssen RH, Rodríguez-Antona C, García-Donas J, Rini BI, Guchelaar HJ. CYP3A5 and ABCB1 Polymorphisms as Predictors for Sunitinib Outcome in Metastatic Renal Cell Carcinoma. Eur Urol 2015; 68:621-9. [DOI: 10.1016/j.eururo.2015.04.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/13/2015] [Indexed: 01/20/2023]
|
86
|
Anderson JC, Willey CD, Mehta A, Welaya K, Chen D, Duarte CW, Ghatalia P, Arafat W, Madan A, Sudarshan S, Naik G, Grizzle WE, Choueiri TK, Sonpavde G. High Throughput Kinomic Profiling of Human Clear Cell Renal Cell Carcinoma Identifies Kinase Activity Dependent Molecular Subtypes. PLoS One 2015; 10:e0139267. [PMID: 26406598 PMCID: PMC4583516 DOI: 10.1371/journal.pone.0139267] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/09/2015] [Indexed: 01/08/2023] Open
Abstract
Despite the widespread use of kinase-targeted agents in clear cell renal cell carcinoma (CC-RCC), comprehensive kinase activity evaluation (kinomic profiling) of these tumors is lacking. Thus, kinomic profiling of CC-RCC may assist in devising a classification system associated with clinical outcomes, and help identify potential therapeutic targets. Fresh frozen CC-RCC tumor lysates from 41 clinically annotated patients who had localized disease at diagnosis were kinomically profiled using the PamStation®12 high-content phospho-peptide substrate microarray system (PamGene International). Twelve of these patients also had matched normal kidneys available that were also profiled. Unsupervised hierarchical clustering and supervised comparisons based on tumor vs. normal kidney and clinical outcome (tumor recurrence) were performed and coupled with advanced network modeling and upstream kinase prediction methods. Unsupervised clustering analysis of localized CC-RCC tumors identified 3 major kinomic groups associated with inflammation (A), translation initiation (B), and immune response and cell adhesions (C) processes. Potential driver kinases implicated include PFTAIRE (PFTK1), PKG1, and SRC, which were identified in groups A, B, and C, respectively. Of the 9 patients who had tumor recurrence, only one was found in Group B. Supervised analysis showed decreased kinase activity of CDK1 and RSK1-4 substrates in those which progressed compared to others. Twelve tumors with matching normal renal tissue implicated increased PIM's and MAPKAPK's in tumors compared to adjacent normal renal tissue. As such, comprehensive kinase profiling of CC-RCC tumors could provide a functional classification strategy for patients with localized disease and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Joshua C. Anderson
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Christopher D. Willey
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Amitkumar Mehta
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Karim Welaya
- Clinical Oncology Department, University of Alexandria, Alexandria, Egypt
| | - Dongquan Chen
- Division of Preventative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Christine W. Duarte
- Center for Outcomes Research and Evaluation (CORE), Maine Medical Center Research Institute (MMCRI), Portland, Maine, United States of America
| | - Pooja Ghatalia
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Waleed Arafat
- Clinical Oncology Department, University of Alexandria, Alexandria, Egypt
| | - Ankit Madan
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Sunil Sudarshan
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Gurudatta Naik
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - William E. Grizzle
- Division of Anatomic Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Toni K. Choueiri
- Department of Medical Oncology, Kidney Cancer Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Guru Sonpavde
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
87
|
Diekstra MHM, Liu X, Swen JJ, Boven E, Castellano D, Gelderblom H, Mathijssen RHJ, Rodríguez-Antona C, García-Donas J, Rini BI, Guchelaar HJ. Association of single nucleotide polymorphisms in IL8 and IL13 with sunitinib-induced toxicity in patients with metastatic renal cell carcinoma. Eur J Clin Pharmacol 2015; 71:1477-84. [PMID: 26387812 PMCID: PMC4643117 DOI: 10.1007/s00228-015-1935-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 08/31/2015] [Indexed: 12/15/2022]
Abstract
Purpose Earlier, the association of single nucleotide polymorphisms (SNPs) with toxicity and efficacy of sunitinib has been explored in patients with metastatic renal cell carcinoma (mRCC). Recently, additional SNPs have been suggested as potential biomarkers. We investigated these novel SNPs for association with sunitinib treatment outcome in mRCC patients. Methods In this exploratory study, we selected SNPs in genes CYP3A4, NR1I2, POR, IL8, IL13, IL4-R, HIF1A and MET that might possibly be associated with sunitinib treatment outcome. Each SNP was tested for association with progression-free survival (PFS) and overall survival (OS) by Cox-regression analysis and for clinical response and toxicity using logistic regression. Results We included 374 patients for toxicity analyses, of which 38 patients with non-clear cell renal cell cancer were excluded from efficacy analyses. The risk for hypertension was increased in the presence of the T allele in IL8 rs1126647 (OR = 1.69, 95 % CI = 1.07–2.67, P = 0.024). The T allele in IL13 rs1800925 was associated with an increase in the risk of leukopenia (OR = 6.76, 95 % CI = 1.35–33.9, P = 0.020) and increased prevalence of any toxicity > grade 2 (OR = 1.75, 95 % CI = 1.06–2.88, P = 0.028). No significant associations were found with PFS, OS or clinical response. Conclusions We show that polymorphisms in IL8 rs1126647 and IL13 rs1800925 are associated with sunitinib-induced toxicities. Validation in an independent cohort is required. Electronic supplementary material The online version of this article (doi:10.1007/s00228-015-1935-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Meta H M Diekstra
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, Netherlands
- Dutch SUTOX consortium, Leiden, Netherlands
| | - Xiaoyan Liu
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, Netherlands
- Dutch SUTOX consortium, Leiden, Netherlands
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, China
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, Netherlands.
- Dutch SUTOX consortium, Leiden, Netherlands.
| | - Epie Boven
- Dutch SUTOX consortium, Leiden, Netherlands
- Department of Medical Oncology, VU University Medical Center, Amsterdam, Netherlands
| | - Daniel Castellano
- Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Spanish Oncology Genitourinary Group (SOGUG), Madrid, Spain
| | - Hans Gelderblom
- Dutch SUTOX consortium, Leiden, Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Ron H J Mathijssen
- Dutch SUTOX consortium, Leiden, Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Cristina Rodríguez-Antona
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Jesus García-Donas
- Spanish Oncology Genitourinary Group (SOGUG), Madrid, Spain
- Oncology Unit, Clara Campal Comprehensive Cancer Center, Madrid, Spain
| | - Brian I Rini
- Department of Solid Tumor Oncology, Cleveland Clinic Taussig Cancer Institute (CCF), Cleveland, OH, USA
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, Netherlands
- Dutch SUTOX consortium, Leiden, Netherlands
| |
Collapse
|
88
|
Komatsu Y, Ohki E, Ueno N, Yoshida A, Toyoshima Y, Ueda E, Houzawa H, Togo K, Nishida T. Safety, efficacy and prognostic analyses of sunitinib in the post-marketing surveillance study of Japanese patients with gastrointestinal stromal tumor. Jpn J Clin Oncol 2015; 45:1016-22. [PMID: 26373318 PMCID: PMC4622169 DOI: 10.1093/jjco/hyv126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/29/2015] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE This study was conducted to expand the sunitinib safety database in Japanese imatinib-resistant/-intolerant gastrointestinal stromal tumor patients. Retrospective analyses investigated common adverse events as potential prognostic markers. METHODS Four hundred and seventy patients who received sunitinib between June 2008 and November 2009 were analyzed for safety, progression-free survival and overall survival; 386 for objective response rate; 88% received sunitinib on Schedule 4/2 starting at 50 mg/day. RESULTS No unexpected safety issues occurred. Grade ≥ 3 adverse events occurred in 70%, most commonly thrombocytopenia (33%), neutropenia (22%) and leukopenia (15%). Objective response rate was 20% (95% confidence interval 16-24). Median progression-free survival was 22.4 weeks (95% confidence interval, 21.7-24.0). The overall survival rate at 24 weeks was 91% (95% confidence interval, 88-94). Higher relative dose intensity (≥70 vs. <70%) during the first 6 weeks and better Eastern Cooperative Oncology Group performance status (0 vs. ≥1) were associated with longer progression-free survival (24.0 vs. 20.1 weeks; P = 0.011; and 24.1 vs. 16.9 weeks; P < 0.001) and higher 24-week overall survival rate (94 vs. 83%; P < 0.001; and 96 vs. 83%; P < 0.001). Increased progression-free survival and overall survival rates were associated with specific adverse events. Cox proportional hazard modeling adjusted for relative dose intensity and performance status established hand-foot syndrome (hazard ratio = 0.636; 95% confidence interval, 0.456-0.888) and leukopenia (hazard ratio = 0.683; 95% confidence interval, 0.492-0.948) occurring within 12 weeks were significantly correlated with increased progression-free survival. CONCLUSION Sunitinib showed good efficacy and tolerable safety. Factors associated with greater efficacy were relative dose intensity, performance status and specific early adverse events.
Collapse
Affiliation(s)
- Yoshito Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Kita-ku, Sapporo
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Pretreatment neutrophil-to-lymphocyte ratio predicts prognosis in patients with metastatic renal cell carcinoma receiving targeted therapy. Int J Clin Oncol 2015; 21:373-378. [PMID: 26335242 DOI: 10.1007/s10147-015-0894-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 08/16/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND The neutrophil-to-lymphocyte ratio (NLR) is associated with clinical outcomes of various cancers. This study aimed to evaluate whether pretreatment NLR can be used as a prognostic factor in patients with metastatic renal cell carcinoma (mRCC) receiving targeted therapy. METHODS In this single-center retrospective study, the Kaplan-Meier method was used to estimate progression-free survival (PFS) and overall survival (OS) of 373 mRCC patients receiving targeted therapy. The survival outcomes of patients with high (≥ 2.2) and low (< 2.2) pretreatment NLRs were compared by log-rank test, and Cox proportional hazard regression model was used to compare OS and PFS between groups. RESULTS The overall median PFS and OS times for all 373 patients were 18.4 and 34.3 months, respectively. Patients with high NLRs had significantly shorter median OS (28.8 vs 410 months, P = 0.005) and PFS (15.4 vs 23.9 months, P = 0.001) than those with low NLRs. After adjusting for confounding variables, each unit increase of NLR was associated with a 40 % increase in mortality (hazard ratio [HR] 1.391; 95 % confidence interval [CI] 1.022-1.894; P = 0.036). High NLR was also an independent predictor of poor PFS (HR 1.544; 95 % CI 1.166-2.045; P = 0.002). CONCLUSION Pretreatment NLR may be an independent prognostic factor for mRCC patients who are receiving targeted therapy.
Collapse
|
90
|
Chen J, Lu Y, Zheng Y. Incidence and risk of hypertension with bevacizumab in non-small-cell lung cancer patients: a meta-analysis of randomized controlled trials. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:4751-60. [PMID: 26316712 PMCID: PMC4547635 DOI: 10.2147/dddt.s87258] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aim A study was conducted to determine the overall risk and incidence of hypertension with bevacizumab in non-small-cell lung cancer (NSCLC) patients. Materials and methods Electronic databases such as the Embase, PubMed, and Cochrane Library were searched for related trials. Statistical analyses were conducted to calculate the overall incidence rates, odds ratios (ORs), and 95% confidence intervals (CIs) by using either random-effect or fixed-effect models depending on the heterogeneity. Results A total of 3,155 subjects from nine studies were included. The overall incidences of all-grade and high-grade hypertension in NSCLC patients were 19.55% (95% CI 10.17%–34.3%) and 6.95% (95% CI 5.81%–8.30%). Bevacizumab use was associated with a significantly increased risk in all-grade hypertension (OR 8.07, 95% CI 3.87–16.85; P=0.0002) and high-grade hypertension (OR 5.93, 95% CI 3.41–10.32; P<0.0001). No evidence of publication bias was determined for the ORs of hypertension in our meta-analysis. Conclusion Bevacizumab is associated with a significantly increased risk of hypertension development in NSCLC patients. Early monitoring and effective management of hypertension might be important steps for the safe use of this drug.
Collapse
Affiliation(s)
- Jian Chen
- Intensive Care Unit, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yingfeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yunliang Zheng
- Research Center for Clinical Pharmacy, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
91
|
Berardi R, Brunelli A, Pagliaretta S, Paolucci V, Conti A, Goteri G, Refai M, Pompili C, Marcantognini G, Morgese F, Ballatore Z, Savini A, De Lisa M, Caramanti M, Santoni M, Zizzi A, Piva F, Mazzanti P, Onofri A, Sabbatini A, Scarpelli M, Cascinu S. Impact of VEGF, VEGFR, PDGFR, HIF and ERCC1 gene polymorphisms on thymic malignancies outcome after thymectomy. Oncotarget 2015; 6:19305-19315. [PMID: 26254278 PMCID: PMC4662492 DOI: 10.18632/oncotarget.4191] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 05/26/2015] [Indexed: 02/06/2023] Open
Abstract
We aimed to analyze genotypes of VEGF-A, VEGFR2, Flt4, PDGFRα, HIF-1α and ERCC1 and their correlation with thymic tumor risk and patient outcome. DNA of 57 consecutive patients (43 thymomas and 14 thymic carcinomas) who underwent total thymectomy at our Institution was extracted from paraffin-embedded tissue. We selected polymorphisms in the following genes:HIF1-α (rs2057482T > C, rs1951795A > C, rs2301113C > A, rs10873142C > T, rs11158358G > C, rs12434438G > A, rs11549465C > T, rs11549467G > A), VEGF-A (rs2010963G > C, rs699947A > C), VEGFR-2 (rs2305948C > T, rs1870377T > A), VEGFR-3 (rs307826T > C, rs307821C > A), PDGFR-α (rs35597368C > T) and ERCC1 (rs11615A > G). Gene polymorphisms were determined by Real-Time PCR using TaqMan assays. As compared to the general population, the allele frequency of PDGFR-α rs35597368T was significantly higher (95% vs. 87%, p = 0.036), while the frequency of alleles HIF1-α rs2057482C (78% vs. 90%), rs1951795C (69% vs. 87%), rs2301113A (70% vs. 83%), rs10873142T (70% vs. 87%), rs11158358C (75% vs. 88%), rs12434438A (67% vs. 84%) were significantly lower. VEGFR-3 rs307821C frequency was significantly higher in thymomas vs. thymic carcinomas (79% vs. 72%, p = 0.0371). The following factors were significantly correlated with a longer overall survival: VEGFR-3 rs307826C, VEGFR-2 rs1870377A, PDGFR-α rs35597368T/C, HIF1-α rs2301113C, rs2057482C/T, rs1951795C, rs11158358G/C and rs10873142T/C, ERCC1 rs11615A (p < 0.05). Our results suggest, for the first time, that PDGFR-α, HIF-1α and VEGFR-3 SNPs are associated with thymic cancer risk and survival.
Collapse
Affiliation(s)
- Rossana Berardi
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Alessandro Brunelli
- Department of Thoracic Surgery, St. James's University Hospital, Leeds, West Yorkshire, United Kingdom
| | - Silvia Pagliaretta
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Vittorio Paolucci
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | | | - Gaia Goteri
- Section of Pathological Anatomy and Histopathology, Università Politecnica delle Marche, Ancona, Italy
| | - Majed Refai
- Thoracic Surgery, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Cecilia Pompili
- Department of Thoracic Surgery, St. James's University Hospital, Leeds, West Yorkshire, United Kingdom
| | - Giulia Marcantognini
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Francesca Morgese
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Zelmira Ballatore
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Agnese Savini
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Mariagrazia De Lisa
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Miriam Caramanti
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Matteo Santoni
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Antonio Zizzi
- Section of Pathological Anatomy and Histopathology, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Paola Mazzanti
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Azzurra Onofri
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Armando Sabbatini
- Thoracic Surgery, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| | - Marina Scarpelli
- Section of Pathological Anatomy and Histopathology, Università Politecnica delle Marche, Ancona, Italy
| | - Stefano Cascinu
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I – GM Lancisi – G Salesi, Italy
| |
Collapse
|
92
|
Chu YH, Li H, Tan HS, Koh V, Lai J, Phyo WM, Choudhury Y, Kanesvaran R, Chau NM, Toh CK, Ng QS, Tan PH, Chowbay B, Tan MH. Association of ABCB1 and FLT3 Polymorphisms with Toxicities and Survival in Asian Patients Receiving Sunitinib for Renal Cell Carcinoma. PLoS One 2015; 10:e0134102. [PMID: 26244574 PMCID: PMC4526634 DOI: 10.1371/journal.pone.0134102] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/06/2015] [Indexed: 11/26/2022] Open
Abstract
Sunitinib is a tyrosine kinase inhibitor used as first-line treatment for metastatic renal cell carcinoma (mRCC). Asian ethnicity has been previously associated with lower clearance and greater toxicities for sunitinib treatment, relative to Caucasian ethnicity. Research focusing on identifying corresponding biomarkers of efficacy and toxicity has been hitherto conducted in Caucasian populations, and few of the reported associations have been externally validated. Our work thus aims to investigate candidate biomarkers in Asian patients receiving sunitinib, comparing the observed genotype effects with those reported in Caucasian populations. Using data from 97 Asian mRCC patients treated with sunitinib, we correlated 7 polymorphisms in FLT3, ABCB1, VEGFR2, ABCG2 and BIM with patient toxicities, response, and survival. We observed a stronger association of FLT3 738T genotype with leucopenia in our Asian dataset than that previously reported in Caucasian mRCC patients (odds ratio [OR]=8.0; P=0.03). We observed significant associations of FLT3 738T (OR=2.7), ABCB1 1236T (OR=0.3), ABCB1 3435T (OR=0.1), ABCB1 2677T (OR=0.4), ABCG2 421A (OR=0.3) alleles and ABCB1 3435, 1236, 2677 TTT haplotype (OR=0.1) on neutropenia. Primary resistance (OR=0.1, P=0.004) and inferior survival (progression-free: hazard ratio [HR]=5.5, P=0.001; overall: HR=5.0, P=0.005) were associated with the ABCB1 3435, 1236, 2677 TTT haplotype. In conclusion, ABCB1 and FLT3 polymorphisms may be helpful in predicting sunitinib toxicities, response and survival benefit in Asian mRCC patients. We have also validated the association between FLT3 738T and sunitinib-induced leucopenia previously reported in Caucasian populations, but have not validated other reported genetic associations.
Collapse
Affiliation(s)
- Ying-Hsia Chu
- Institute of Bioengineering and Nanotechnology, Singapore, Republic of Singapore
| | - Huihua Li
- Health Services Research, Singapore General Hospital, Singapore, Republic of Singapore
| | - Hui Shan Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Valerie Koh
- Department of Pathology, Singapore General Hospital, Singapore, Republic of Singapore
| | - Johnathan Lai
- Institute of Bioengineering and Nanotechnology, Singapore, Republic of Singapore
| | - Wai Min Phyo
- Institute of Bioengineering and Nanotechnology, Singapore, Republic of Singapore
| | - Yukti Choudhury
- Institute of Bioengineering and Nanotechnology, Singapore, Republic of Singapore
| | - Ravindran Kanesvaran
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Noan Minh Chau
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Chee Keong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Quan Sing Ng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Puay Hoon Tan
- Department of Pathology, Singapore General Hospital, Singapore, Republic of Singapore
| | - Balram Chowbay
- Laboratory of Clinical Pharmacology, Division Medical Sciences, National Cancer Centre Singapore, Singapore, Republic of Singapore
- Clinical Pharmacology Core, SingHealth, Singapore, Republic of Singapore
- Clinical Sciences, Duke-NUS Graduate Medical School, Singapore, Republic of Singapore
| | - Min-Han Tan
- Institute of Bioengineering and Nanotechnology, Singapore, Republic of Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Republic of Singapore
- * E-mail:
| |
Collapse
|
93
|
Predictive biomarker candidates to delineate efficacy of antiangiogenic treatment in renal cell carcinoma. Clin Transl Oncol 2015; 18:1-8. [PMID: 26169213 DOI: 10.1007/s12094-015-1332-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/18/2015] [Indexed: 02/07/2023]
Abstract
Antiangiogenic therapy is currently considered as the cornerstone of treatment in metastatic kidney cancer. A monoclonal antibody against the vascular endothelial growth factor (VEGF) and several tyrosine kinase inhibitors targeting the VEGF receptors demonstrated, 7 years ago, to deeply impact the outcome of this tumor and became a model of integration of molecular knowledge into clinical practice. Unfortunately, no further improvement in survival has been made and 20-25 % of cases remain primary refractory to these drugs, with an overall dismal prognosis. Since biomarker predictors of activity are lacking, their development could highly help in the process of making clinical decisions when choosing the best option for every patient or prompting the inclusion in clinical trials. This unmet medical need could become even more relevant if new immunotherapy confirms its initial promising results in this pathology. In this article, we provide an insight of current state of the art regarding the prediction of antiangiogenic efficacy in kidney cancer and propose new strategies for the implementation of such markers in clinical practice.
Collapse
|
94
|
|
95
|
Grande E, Capdevila J, Castellano D, Teulé A, Durán I, Fuster J, Sevilla I, Escudero P, Sastre J, García-Donas J, Casanovas O, Earl J, Ortega L, Apellaniz-Ruiz M, Rodriguez-Antona C, Alonso-Gordoa T, Díez JJ, Carrato A, García-Carbonero R. Pazopanib in pretreated advanced neuroendocrine tumors: a phase II, open-label trial of the Spanish Task Force Group for Neuroendocrine Tumors (GETNE). Ann Oncol 2015; 26:1987-1993. [PMID: 26063633 DOI: 10.1093/annonc/mdv252] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/26/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The management of advanced neuroendocrine tumors (NETs) has recently changed. We assessed the activity of pazopanib after failure of other systemic treatments in advanced NETs. METHODS This was a multicenter, open-label, phase II study evaluating pazopanib as a single agent in advanced NETs (PAZONET study). The clinical benefit rate (CBR) at 6 months was the primary end point. Translational correlation of radiological response and progression-free survival (PFS) with circulating and tissue biomarkers was also evaluated. RESULTS A total of 44 patients were enrolled. Twenty-five patients (59.5%) were progression-free at 6 months (4 partial responses, 21 stable diseases) with a median PFS of 9.5 months [95% confidence interval (CI) 4.8-14.1]. The CBR varied according to prior therapy received, with 73%, 60% and 25% in patients treated with prior multitarget inhibitors, prior mTOR inhibitors and both agents, respectively. A nonsignificant increase in PFS was observed in patients presenting lower baseline circulating tumor cell (CTC) counts (9.1 versus 5.8 months; P = 0.22) and in those with decreased levels of soluble-vascular endothelial growth factor receptor-2 (sVEGFR-2) (12.6 versus 9.1 months; P = 0.067). A trend toward reduced survival was documented in patients with VEGFR3 rs307821 and rs307826 missense polymorphisms [hazard ratio (HR): 12.3; 95% CI 1.09-139.2; P = 0.042 and HR: 6.9; 95% CI 0.96-49.9; P = 0.055, respectively]. CONCLUSIONS Pazopanib showed clinical activity in patients with advanced NETs regardless of previous treatments. Additionally, CTCs, soluble-s VEFGR-2 and VEGFR3 gene polymorphisms constitute potential biomarkers for selecting patients for pazopanib (NCT01280201). CLINICAL TRIAL NUMBER NCT01280201.
Collapse
Affiliation(s)
- E Grande
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid.
| | - J Capdevila
- Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona
| | - D Castellano
- Department of Medical Oncology, I + 12 Research Institute, 12 de Octubre University Hospital, Madrid
| | - A Teulé
- Department of Medical Oncology, IDIBELL, Catalan Institute of Oncology L'Hospitalet, Barcelona
| | - I Durán
- Department of Medical Oncology, Instituto de Biomedicina de Sevilla (IBIS) [HUVR, CSIC, University of Seville], Virgen del Rocío University Hospital, Seville
| | - J Fuster
- Department of Medical Oncology, Son Espases Hospital, Palma de Mallorca
| | - I Sevilla
- Department of Medical Oncology, Virgen de la Victoria University Hospital, Malaga
| | - P Escudero
- Department of Medical Oncology, Clínico Lozano Blesa University Hospital, Zaragoza
| | - J Sastre
- Department of Medical Oncology, Clínico San Carlos Hospital, Madrid
| | - J García-Donas
- Department of Medical Oncology, Centro Integral Oncológico Clara Campal, Madrid
| | - O Casanovas
- Tumor Angiogenesis Group, IDIBELL, Catalan Institute of Oncology L'Hospitalet, Barcelona
| | - J Earl
- Department of Medical Oncology Research Laboratory, Ramón y Cajal University Hospital, Madrid
| | - L Ortega
- Department of Pathology, Clínico San Carlos Hospital, Madrid
| | - M Apellaniz-Ruiz
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center, ISCIII Center for Biomedical Research on Rare Disease (CIBERER) Madrid, Madrid
| | - C Rodriguez-Antona
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center, ISCIII Center for Biomedical Research on Rare Disease (CIBERER) Madrid, Madrid
| | - T Alonso-Gordoa
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid
| | - J J Díez
- Department of Endocrinology, Ramón y Cajal University Hospital, Madrid, Spain
| | - A Carrato
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid
| | - R García-Carbonero
- Department of Medical Oncology, Instituto de Biomedicina de Sevilla (IBIS) [HUVR, CSIC, University of Seville], Virgen del Rocío University Hospital, Seville
| |
Collapse
|
96
|
Precision medicine in oncology needs to integrate pharmacogenetic profiling. Eur Urol 2015; 68:630-1. [PMID: 26044803 DOI: 10.1016/j.eururo.2015.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 05/20/2015] [Indexed: 11/20/2022]
|
97
|
Takayoshi K, Sagara K, Uchino K, Kusaba H, Sakamoto N, Iguchi A, Baba E. A case of metastatic renal cell carcinoma and bile duct carcinoma treated with a combination of sunitinib and gemcitabine. BMC Cancer 2015; 15:426. [PMID: 26001650 PMCID: PMC4491890 DOI: 10.1186/s12885-015-1443-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 05/15/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Metastatic renal cell carcinoma (mRCC) had been a chemo-refractory disease, but recent advances in multiple kinase inhibitors such as sunitinib have dramatically changed the clinical course of mRCC. Sunitinib is used for mRCC chemotherapy based on the favorable results of a recent clinical trial, but specific biomarkers predicting efficacy and safety are not yet available. Locally advanced bile duct carcinoma (BDC) has generally been treated with single agent gemcitabine or as doublet therapy with cisplatin. Concomitant occurrence of mRCC and BDC is extremely rare, and a standard therapeutic strategy has not been established. CASE PRESENTATION A 65-year-old woman was diagnosed as having multiple mRCC and intercurrent, locally advanced BDC. A single course of combination therapy with sunitinib (25 mg/day, day2-15) and gemcitabine (750 mg/m(2), days 1, 8) was administered, and this showed obvious effects, with partial response for mRCC and stable disease for BDC. However, the patient also experienced severe adverse events, including hematological and various non-hematological toxicities; the combination therapy was then terminated on day 13 after its initiation. She recovered on day 28 and is alive 3.5 years after the diagnosis. The plasma trough levels of sunitinib and its active metabolite SU12662 on day 13 were 91.5 ng/mL and 19.2 ng/mL, respectively, which were relatively higher than in previous reports. Analysis of her single nucleotide polymorphisms (SNPs) detected TC in ABCB1 3435C/T, TC in 1236C/T and TT in 2677G/T, suggesting a possible TTT haplotype. CONCLUSION A rare case of double cancer of mRCC and BDC was treated by combination chemotherapy. Although unknown synergistic mechanisms of these agents may be involved, severe toxicities might be possibly associated with high sunitinib exposure. Further exploration of combination therapy with sunitinib and gemcitabine is required.
Collapse
Affiliation(s)
- Kotoe Takayoshi
- Department of Medical Oncology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, 1-8-1 Jigyouhama, Chuo-ku, Fukuoka, 810-8563, Japan.
| | - Kosuke Sagara
- Department of Medical Oncology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, 1-8-1 Jigyouhama, Chuo-ku, Fukuoka, 810-8563, Japan.
| | - Keita Uchino
- Department of Medical Oncology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, 1-8-1 Jigyouhama, Chuo-ku, Fukuoka, 810-8563, Japan.
| | - Hitoshi Kusaba
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.
| | - Naotaka Sakamoto
- Department of Urology, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan.
| | - Atsushi Iguchi
- Department of Urology, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan.
| | - Eishi Baba
- Department of Comprehensive Clinical Oncology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
98
|
NCCTG N0821 (Alliance): a phase II first-line study of pemetrexed, carboplatin, and bevacizumab in elderly patients with advanced nonsquamous non-small-cell lung cancer with good performance status. J Thorac Oncol 2015; 9:1146-53. [PMID: 25157767 DOI: 10.1097/jto.0000000000000217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND We hypothesized that the combination of bevacizumab, carboplatin, and pemetrexed will be an effective first-line regimen in fit, elderly patients with nonsquamous non-small-cell lung cancer. METHODS Treatment-naïve, stage IIIB/IV nonsquamous non-small-cell lung cancer patients more than 70 years old with good performance status (Eastern Cooperative Oncology Group performance status 0-1) and adequate organ function were eligible. Carboplatin area under the curve 6, pemetrexed 500 mg/m, and bevacizumab 15 mg/kg were administered on day 1 of each 21-day cycle (up to six cycles) followed by maintenance pemetrexed and bevacizumab. The primary end point of 6-month progression-free survival rate of at least 70% was assessed using a one-stage binomial design. Quality of life (QOL) questionnaires were administered. Polymorphisms in genes encoding relevant proteins (drug targets, transport, and metabolism proteins) were correlated with treatment outcome. RESULTS Fifty-seven eligible patients were enrolled. Median age was 74.5 years. Median treatment cycles received was 6. The most common grade 3 or higher non-hematologic adverse events were fatigue (26%) and hypertension (11%); 16% had grade 4 neutropenia and 6.5% had grade 4 thrombocytopenia. Three patients experienced grade 3/4 hemorrhagic events (one pulmonary, two gastrointestinal). Primary end point of PFS6 was 60% (95% confidence interval [CI]: 45.9-73%). Median PFS was 7.0 months (95% CI: 5.9-10.1), median overall survival was 13.7 months (95% CI: 9.4-16.8). Polymorphic KDR and VEGFA variants correlated with survival and toxicity, respectively. There was no significant change in overall QOL scores over time. CONCLUSION This regimen is feasible and did not decrease the QOL in this study population. However, it did not meet the primary efficacy end point.
Collapse
|
99
|
Terada T, Noda S, Inui KI. Management of dose variability and side effects for individualized cancer pharmacotherapy with tyrosine kinase inhibitors. Pharmacol Ther 2015; 152:125-34. [PMID: 25976912 DOI: 10.1016/j.pharmthera.2015.05.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/23/2015] [Indexed: 12/18/2022]
Abstract
Molecular-targeted therapies with tyrosine kinase inhibitors (TKIs) have provided a major breakthrough in cancer treatment. These agents are given orally and demonstrated to be substrates for drug transporters. In clinical settings, TKIs are mainly used at a fixed dose, but wide interpatient variability has been observed in their pharmacokinetics and/or pharmacodynamics. Genetic polymorphisms of ABC transporters, drug-drug interaction and adherence are among the factors causing such variation. To overcome these problems, therapeutic drug monitoring has been applied in clinical practice for patient care. Skin disorders are frequently observed as adverse drug reactions when using TKIs, and are commonly managed by symptomatic therapy based on clinical experience. Recent studies have provided some insights into the molecular mechanisms underlying skin disorders induced by TKIs. This review article summarizes the accumulated clinical and basic pharmacological evidence of TKIs, focusing on erlotinib, sorafenib and sunitinib.
Collapse
Affiliation(s)
- Tomohiro Terada
- Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu City, Shiga 520-2192, Japan.
| | - Satoshi Noda
- Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu City, Shiga 520-2192, Japan
| | - Ken-Ichi Inui
- Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8414, Kyoto, Japan
| |
Collapse
|
100
|
Chau CH, Price DK, Till C, Goodman PJ, Chen X, Leach RJ, Johnson-Pais TL, Hsing AW, Hoque A, Tangen CM, Chu L, Parnes HL, Schenk JM, Reichardt JKV, Thompson IM, Figg WD. Finasteride concentrations and prostate cancer risk: results from the Prostate Cancer Prevention Trial. PLoS One 2015; 10:e0126672. [PMID: 25955319 PMCID: PMC4425512 DOI: 10.1371/journal.pone.0126672] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 04/06/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE In the Prostate Cancer Prevention Trial (PCPT), finasteride reduced the risk of prostate cancer by 25%, even though high-grade prostate cancer was more common in the finasteride group. However, it remains to be determined whether finasteride concentrations may affect prostate cancer risk. In this study, we examined the association between serum finasteride concentrations and the risk of prostate cancer in the treatment arm of the PCPT and determined factors involved in modifying drug concentrations. METHODS Data for this nested case-control study are from the PCPT. Cases were drawn from men with biopsy-proven prostate cancer and matched controls. Finasteride concentrations were measured using a liquid chromatography-mass spectrometry validated assay. The association of serum finasteride concentrations with prostate cancer risk was determined by logistic regression. We also examine whether polymorphisms in the enzyme target and metabolism genes of finasteride are related to drug concentrations using linear regression. RESULTS AND CONCLUSIONS Among men with detectable finasteride concentrations, there was no association between finasteride concentrations and prostate cancer risk, low-grade or high-grade, when finasteride concentration was analyzed as a continuous variable or categorized by cutoff points. Since there was no concentration-dependent effect on prostate cancer, any exposure to finasteride intake may reduce prostate cancer risk. Of the twenty-seven SNPs assessed in the enzyme target and metabolism pathway, five SNPs in two genes, CYP3A4 (rs2242480; rs4646437; rs4986910), and CYP3A5 (rs15524; rs776746) were significantly associated with modifying finasteride concentrations. These results suggest that finasteride exposure may reduce prostate cancer risk and finasteride concentrations are affected by genetic variations in genes responsible for altering its metabolism pathway. TRIAL REGISTRATION ClinicalTrials.gov NCT00288106.
Collapse
Affiliation(s)
- Cindy H. Chau
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Douglas K. Price
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Cathee Till
- Swog Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Phyllis J. Goodman
- Swog Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Xiaohong Chen
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Robin J. Leach
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Teresa L. Johnson-Pais
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Ann W. Hsing
- Cancer Prevention Institute of California, Fremont, California, Stanford Cancer Institute, Palo Alto, California, United States of America
| | - Ashraful Hoque
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Catherine M. Tangen
- Swog Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lisa Chu
- Cancer Prevention Institute of California, Fremont, California, Stanford Cancer Institute, Palo Alto, California, United States of America
| | - Howard L. Parnes
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jeannette M. Schenk
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Juergen K. V. Reichardt
- School of Pharmacy and Molecular Sciences, James Cook University, Townsville, Queensland, Australia
| | - Ian M. Thompson
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - William D. Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|