1
|
Feng W, Zhou L, He J, Wang Y, Cai J, Jiang T, Zhao Q, Ren T. Association of VEGFR2 polymorphisms with clinical outcomes of anti-angiogenesis therapy in cancer patients: A systematic review and meta-analysis. Eur J Pharmacol 2025; 990:177299. [PMID: 39864574 DOI: 10.1016/j.ejphar.2025.177299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/02/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Some cancer patients derive limited benefit from anti-angiogenic therapy or discontinuation due to adverse reactions. Vascular endothelial growth factor receptor 2 (VEGFR2) plays an important role in regulating angiogenesis in tumors. This study aims to evaluate the association of VEGFR2 polymorphisms with clinical outcomes of anti-angiogenic drugs (AADs) in cancer patients. METHODS PubMed, Embase, Web of Science, and the Cochrane Library were searched from inception to Dec 26, 2023. Studies accessing the association of VEGFR2 polymorphisms with efficacy and/or safety of AADs in patients with solid tumor were included. RESULTS A total of 32 studies encompassing 7075 patients were identified. The T allele of rs2305948 (C > T) was significantly associated with worse progression-free survival and overall survival, especially in Asians, patients with the dominant model (CT/TT vs. CC), bevacizumab-treated patients, colorectal cancer patients, and non-small cell lung cancer patients. The C allele of rs2071559 (T > C) was markedly associated with worse PFS and OS, specifically in the dominant model (CC/CT vs. TT), apatinib-treated patients, and non-small cell lung cancer patients. The A allele of rs1870377 (T > A) was significantly associated with improved PFS, particularly in patients with renal cell carcinoma. However, this A allele also significantly increased the risk of hypertension. No significant associations were observed for rs2305948 (G > A), rs11133360 (T > C), and rs12505758 (T > C) with the clinical outcomes of AADs. CONCLUSION Among VEGFR2 polymorphisms, rs2305948 (C > T) and rs2071559 (T > C) were associated with a high risk of disease progression and death, rs1870377 (T > A) was associated with improved PFS but an increased risk of hypertension.
Collapse
Affiliation(s)
- Wenzheng Feng
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Lijun Zhou
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Junyao He
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yimin Wang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jiali Cai
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China; School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Tianhao Jiang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China; School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Qingchun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Tianshu Ren
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
2
|
Numakura K, Igarashi R, Takahashi M, Nara T, Kanda S, Saito M, Narita S, Inoue T, Niioka T, Miura M, Habuchi T. Influence of genetic polymorphisms in vascular endothelial-related genes on the clinical outcome of axitinib in patients with metastatic renal cell carcinoma. Cancer Biol Ther 2024; 25:2312602. [PMID: 38327067 PMCID: PMC10857686 DOI: 10.1080/15384047.2024.2312602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 01/28/2024] [Indexed: 02/09/2024] Open
Abstract
OBJECTIVE Axitinib is an oral multi-target tyrosine kinase inhibitor used for the treatment of renal cell carcinoma (RCC). Because of the severe adverse events (AEs) associated with axitinib, patients often need dose reductions or discontinue its use, highlighting the need for effective biomarkers to assess efficacy and/or AEs. The aim of this study was to investigate the relationship between single nucleotide polymorphisms (SNPs) in genes involved in the pharmacodynamic action of axitinib and clinical prognosis and AEs in metastatic RCC (mRCC) patients. METHODS This study included 80 mRCC patients treated with first-, second-, or third-line axitinib (5 mg orally twice daily). Clinical parameters and genetic polymorphisms were examined in 75 cases (53 males and 22 females). We assessed three SNPs in each of three candidate genes namely, angiotensin-converting enzyme (ACE), nitric oxide synthase 3 (NOS3), and angiotensin II receptor type 1 (AT1R), all of which are involved in axitinib effects on vascular endothelial function. RESULTS Axitinib-treated patients carrying the ACE deletion allele suffered more frequently from hand-foot syndrome and a deterioration in kidney function (p = .045 and p = 0.005, respectively) whereas those carrying the NOS3 G allele suffered more frequently from proteinuria and multiple AEs (p = .025 and p = 0.036, respectively). CONCLUSIONS Our study found that the ACE deletion allele and the NOS3 G allele are associated with increased AEs.
Collapse
Affiliation(s)
- Kazuyuki Numakura
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Ryoma Igarashi
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Makoto Takahashi
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Taketoshi Nara
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Sohei Kanda
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Mitsuru Saito
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Shintaro Narita
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Takamitsu Inoue
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Takenori Niioka
- Department of Pharmacy, Hirosaki University Hospital, Hirosaki, Japan
| | - Masatomo Miura
- Department of Pharmacy, Akita University Hospital, Akita, Japan
| | - Tomonori Habuchi
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
3
|
Yan W, Hou N, Zheng J, Zhai W. Predictive genomic biomarkers of therapeutic effects in renal cell carcinoma. Cell Oncol (Dordr) 2023; 46:1559-1575. [PMID: 37223875 DOI: 10.1007/s13402-023-00827-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND In recent years, there have been great improvements in the therapy of renal cell carcinoma. Nevertheless, the therapeutic effect varies significantly from person to person. To discern the effective treatment for different populations, predictive molecular biomarkers in response to target, immunological, and combined therapies are widely studied. CONCLUSION This review summarized those studies from three perspectives (SNPs, mutation, and expression level) and listed the relationship between biomarkers and therapeutic effect, highlighting the great potential of predictive molecular biomarkers in metastatic RCC therapy. However, due to a series of reasons, most of these findings require further validation.
Collapse
Affiliation(s)
- Weijie Yan
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Naiqiao Hou
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junhua Zheng
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
4
|
Brackenier C, Kinget L, Cappuyns S, Verslype C, Beuselinck B, Dekervel J. Unraveling the Synergy between Atezolizumab and Bevacizumab for the Treatment of Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:348. [PMID: 36672297 PMCID: PMC9856647 DOI: 10.3390/cancers15020348] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/24/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) with antiangiogenic properties, such as sorafenib, have been the standard choice to systemically treat hepatocellular carcinoma for over a decade. More recently, encouraging results were obtained using immune checkpoint inhibitors, although head-to-head comparisons with sorafenib in phase 3 trials could not demonstrate superiority in terms of overall survival. The IMbrave150 was a breakthrough study that resulted in atezolizumab/bevacizumab, a combination of an antiangiogenic and an immune checkpoint inhibitor, as a new standard of care for advanced HCC. This review discusses the mode of action, clinical efficacy, and biomarker research for both drug classes and for the combination therapy. Moreover, the synergy between atezolizumab and bevacizumab is highlighted, unraveling pathophysiological mechanisms underlying an enhanced anticancer immunity by changing the immunosuppressed to a more immunoreactive tumor microenvironment (TME). This is achieved by upregulation of antigen presentation, upregulation of T-cell proliferation, trafficking and infiltration, impairing recruitment, and proliferation of immunosuppressive cells in the TME. However, more insights are needed to identify biomarkers of response that may improve patient selection and outcome.
Collapse
Affiliation(s)
- Cedric Brackenier
- Department of Gastro-Enterology and Hepatology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Lisa Kinget
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Sarah Cappuyns
- Department of Gastro-Enterology and Hepatology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Digestive Oncology, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Chris Verslype
- Department of Gastro-Enterology and Hepatology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Digestive Oncology, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Benoit Beuselinck
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Jeroen Dekervel
- Department of Gastro-Enterology and Hepatology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Digestive Oncology, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
5
|
Sekino Y, Teishima J, Liang G, Hinata N. Molecular mechanisms of resistance to tyrosine kinase inhibitor in clear cell renal cell carcinoma. Int J Urol 2022; 29:1419-1428. [PMID: 36122306 PMCID: PMC10087189 DOI: 10.1111/iju.15042] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/25/2022] [Indexed: 12/24/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cell carcinoma (RCC). Loss of von Hippel-Lindau tumor suppressor gene is frequently observed in ccRCC and increases the expression of hypoxia-inducible factors and their targets, including epidermal growth factor, vascular endothelial growth factor, and platelet-derived growth factor. Tyrosine kinase inhibitors (TKIs) offer a survival benefit in metastatic renal cell carcinoma (mRCC). Recently, immune checkpoint inhibitors have been introduced in mRCC. Combination therapy with TKIs and immune checkpoint inhibitors significantly improved patient outcomes. Therefore, TKIs still play an essential role in mRCC treatment. However, the clinical utility of TKIs is compromised when primary and acquired resistance are encountered. The mechanism of resistance to TKI is not fully elucidated. Here, we comprehensively reviewed the molecular mechanisms of resistance to TKIs and a potential strategy to overcome this resistance. We outlined the involvement of angiogenesis, non-angiogenesis, epithelial-mesenchymal transition, activating bypass pathways, lysosomal sequestration, non-coding RNAs, epigenetic modifications and tumor microenvironment factors in the resistance to TKIs. Deep insight into the molecular mechanisms of resistance to TKIs will help to better understand the biology of RCC and can ultimately help in the development of more effective therapies.
Collapse
Affiliation(s)
- Yohei Sekino
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Department of Urology, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Jun Teishima
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Gangning Liang
- Department of Urology, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Nobuyuki Hinata
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
6
|
Tan T, Han G, Cheng Z, Jiang J, Zhang L, Xia Z, Wang X, Xia Q. Genetic Polymorphisms in CYP2C19 Cause Changes in Plasma Levels and Adverse Reactions to Anlotinib in Chinese Patients With Lung Cancer. Front Pharmacol 2022; 13:918219. [PMID: 35814206 PMCID: PMC9257029 DOI: 10.3389/fphar.2022.918219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Anlotinib is a small molecular multi-targeting tyrosine kinase inhibitor. Growing evidence indicates that treatment efficacy, and toxicity varies considerably between individuals. Therefore, this study aimed to investigate the relationship between cytochrome P450 (CYP450) gene polymorphisms, drug concentrations, and their adverse reactions in anlotinib-treated patients with lung cancer.Methods: We enrolled 139 patients with lung cancer, treated with anlotinib. Twenty loci in the following five genes of the CYP450 family were genotyped: CYP450 family 3 subfamily A member 5 (CYP3A5), 3 subfamily A member 4 (CYP3A4), 2 subfamily C member 9 (CYP2C9), 2 subfamily C member 19 (CYP2C19), and 1 subfamily A member 2 (CYP1A2). Data on adverse reactions were collected from patients, and plasma anlotinib concentrations were measured.Results: There were significant variances in plasma trough concentration (3.95–52.88 ng/ml) and peak plasma concentration (11.53–42.8 ng/ml) following administration of 8 mg anlotinib. Additionally, there were significant differences in the plasma trough concentration (5.65–81.89 ng/ml) and peak plasma concentration (18.01–107.18 ng/ml) following administration of 12 mg anlotinib. Furthermore, for CYP2C19-rs3814637, the peak plasma concentrations of mutant allele T carriers (TT+CT) were significantly higher than those of wildtypes (CC). For CYP2C19-rs11568732, the peak plasma concentrations of the mutant allele G carriers (GT+GG) were significantly higher than those of the wild-type (TT). More importantly, the incidence rates of hypertension and hemoptysis (peripheral lung cancer) with TT+CT in rs3814637 and GT+GG in rs11568732 were significantly higher than those with CC and TT.Conclusions: The plasma trough and peak concentrations varied significantly for both 8 and 12 mg of anlotinib. Single-nucleotide polymorphisms in CYP2C19 are significantly associated with hypertension, hemoptysis, and anlotinib peak concentrations. Polymorphisms in CYP450 may explain inter-individual differences in anlotinib-related adverse reactions.
Collapse
Affiliation(s)
- Tingfei Tan
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Gongwei Han
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ziwei Cheng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Jiemei Jiang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zitong Xia
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xinmeng Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Quan Xia,
| |
Collapse
|
7
|
Genome-Wide Meta-Analysis Identifies Variants in DSCAM and PDLIM3 That Correlate with Efficacy Outcomes in Metastatic Renal Cell Carcinoma Patients Treated with Sunitinib. Cancers (Basel) 2022; 14:cancers14122838. [PMID: 35740506 PMCID: PMC9220885 DOI: 10.3390/cancers14122838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/24/2022] [Accepted: 06/02/2022] [Indexed: 02/04/2023] Open
Abstract
Individual response to sunitinib in metastatic renal cell carcinoma (mRCC) patients is highly variable. Earlier, sunitinib outcome was related to single nucleotide polymorphisms (SNPs) in CYP3A5 and ABCB1. Our aim is to provide novel insights into biological mechanisms underlying sunitinib action. We included mRCC patients from the European EuroTARGET consortium (n = 550) and the RIKEN cohort in Japan (n = 204) which were analysed separately and in a meta-analysis of genome-wide association studies (GWAS). SNPs were tested for association with progression-free survival (PFS) and overall survival (OS) using Cox regression. Summary statistics were combined using a fixed effect meta-analysis. SNP rs28520013 in PDLIM3 and the correlated SNPs rs2205096 and rs111356738 both in DSCAM, showed genome-wide significance (p < 5 × 10−8) with PFS and OS in the meta-analysis. The variant T-allele of rs28520013 associated with an inferior PFS of 5.1 months compared to 12.5 months in non-carriers (p = 4.02 × 10−10, HR = 7.26). T-allele carriers of rs28520013 showed an inferior OS of 6.9 months versus 30.2 months in non-carriers (p = 1.62 × 10−8, HR = 5.96). In this GWAS we identified novel genetic variants in PDLIM3 and DSCAM that impact PFS and OS in mRCC patients receiving sunitinib. The underlying link between the identified genes and the molecular mechanisms of sunitinib action needs to be elucidated.
Collapse
|
8
|
Antiangiogenic Drug-Induced Proteinuria as a Prognostic Factor in Metastatic Colorectal Cancer. Curr Oncol 2022; 29:3996-4011. [PMID: 35735428 PMCID: PMC9221669 DOI: 10.3390/curroncol29060319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/26/2022] [Accepted: 05/28/2022] [Indexed: 12/14/2022] Open
Abstract
Treatment with bevacizumab is known to cause adverse events such as proteinuria and hypertension, amongst others. However, while bevacizumab-induced hypertension has been linked to increased overall survival (OS), data on proteinuria are controversial. We performed a retrospective analysis to observe the influence of adverse events developed during treatment with bevacizumab and chemotherapy on the OS in patients with metastatic colorectal cancer (mCRC). Kaplan–Meier and log-rank analyses were used to assess differences in OS, and hazard ratios (HR) were estimated using Cox models. Out of the 3497 mCRC patients admitted to our center between 2014 and 2019, 150 met the criteria for inclusion in our analysis. Out of these, 50.7% experienced proteinuria and had reached a longer OS (40 versus 25 months, p = 0.015) and progression-free survival (15 versus 12 months, p = 0.039). The following groups were identified as having a lower risk of death: patients with proteinuria (HR 0.589; 95% CI 0.402–0.863; p = 0.007), one metastatic site (HR 0.533; 95% CI 0.363–0.783; p = 0.001), and non-metastatic stage at diagnosis (HR 0.459; 95% CI 0.293–0.720; p = 0.001). Patients with anemia and diabetes had an increased risk of death. Proteinuria emerges as a useful prognostic factor in mCRC patients undergoing bevacizumab-based systemic therapy, and it could be easily integrated into the decision-making process, thus allowing physicians to further individualize systemic treatments.
Collapse
|
9
|
Burnham EA, Abouda AA, Bissada JE, Nardone-White DT, Beers JL, Lee J, Vergne MJ, Jackson KD. Interindividual Variability in Cytochrome P450 3A and 1A Activity Influences Sunitinib Metabolism and Bioactivation. Chem Res Toxicol 2022; 35:792-806. [PMID: 35484684 PMCID: PMC9131896 DOI: 10.1021/acs.chemrestox.1c00426] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Sunitinib is an orally administered tyrosine kinase inhibitor associated with idiosyncratic hepatotoxicity; however, the mechanisms of this toxicity remain unclear. We have previously shown that cytochromes P450 1A2 and 3A4 catalyze sunitinib metabolic activation via oxidative defluorination leading to a chemically reactive, potentially toxic quinoneimine, trapped as a glutathione (GSH) conjugate (M5). The goals of this study were to determine the impact of interindividual variability in P450 1A and 3A activity on sunitinib bioactivation to the reactive quinoneimine and sunitinib N-dealkylation to the primary active metabolite N-desethylsunitinib (M1). Experiments were conducted in vitro using single-donor human liver microsomes and human hepatocytes. Relative sunitinib metabolite levels were measured by liquid chromatography-tandem mass spectrometry. In human liver microsomes, the P450 3A inhibitor ketoconazole significantly reduced M1 formation compared to the control. The P450 1A2 inhibitor furafylline significantly reduced defluorosunitinib (M3) and M5 formation compared to the control but had minimal effect on M1. In CYP3A5-genotyped human liver microsomes from 12 individual donors, M1 formation was highly correlated with P450 3A activity measured by midazolam 1'-hydroxylation, and M3 and M5 formation was correlated with P450 1A2 activity estimated by phenacetin O-deethylation. M3 and M5 formation was also associated with P450 3A5-selective activity. In sandwich-cultured human hepatocytes, the P450 3A inducer rifampicin significantly increased M1 levels. P450 1A induction by omeprazole markedly increased M3 formation and the generation of a quinoneimine-cysteine conjugate (M6) identified as a downstream metabolite of M5. The nonselective P450 inhibitor 1-aminobenzotriazole reduced each of these metabolites (M1, M3, and M6). Collectively, these findings indicate that P450 3A activity is a key determinant of sunitinib N-dealkylation to the active metabolite M1, and P450 1A (and potentially 3A5) activity influences sunitinib bioactivation to the reactive quinoneimine metabolite. Accordingly, modulation of P450 activity due to genetic and/or nongenetic factors may impact the risk of sunitinib-associated toxicities.
Collapse
Affiliation(s)
- Elizabeth A Burnham
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204, United States
| | - Arsany A Abouda
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204, United States
| | - Jennifer E Bissada
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204, United States
| | - Dasean T Nardone-White
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Jessica L Beers
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Jonghwa Lee
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Matthew J Vergne
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, Tennessee 37204, United States
| | - Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
10
|
Franczyk B, Rysz J, Gluba-Brzózka A. Pharmacogenetics of Drugs Used in the Treatment of Cancers. Genes (Basel) 2022; 13:311. [PMID: 35205356 PMCID: PMC8871547 DOI: 10.3390/genes13020311] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
Pharmacogenomics is based on the understanding of the individual differences in drug use, the response to drug therapy (efficacy and toxicity), and the mechanisms underlying variable drug responses. The identification of DNA variants which markedly contribute to inter-individual variations in drug responses would improve the efficacy of treatments and decrease the rate of the adverse side effects of drugs. This review focuses only on the impact of polymorphisms within drug-metabolizing enzymes on drug responses. Anticancer drugs usually have a very narrow therapeutic index; therefore, it is very important to use appropriate doses in order to achieve the maximum benefits without putting the patient at risk of life-threatening toxicities. However, the adjustment of the appropriate dose is not so easy, due to the inheritance of specific polymorphisms in the genes encoding the target proteins and drug-metabolizing enzymes. This review presents just a few examples of such polymorphisms and their impact on the response to therapy.
Collapse
Affiliation(s)
| | | | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland; (B.F.); (J.R.)
| |
Collapse
|
11
|
Joshi A, Patel I, Kapse P, Singh M. Comparative Evaluation of Safety and Efficacy of Alternate Schedule (AS) of Sunitinib in Asian and Non-Asian Patient Population for the Treatment of Metastatic Renal Cell Cancer (mRCC): A Meta-Analysis. KIDNEY CANCER 2022. [DOI: 10.3233/kca-210122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Treatment of metastatic renal cell carcinoma (mRCC) using traditional schedule (TS, 4/2) of Sunitinib is associated with higher adverse effects compared to the alternate schedule (AS, 2/1 upfront or when switched from TS). Objective: This meta-analysis aims to compare the safety, efficacy, and percentage of patients requiring dose reduction or dose interruption between Asian (AP) and non-Asian population (NAP) receiving AS of sunitinib. Methods: Electronic databases (PubMed, EMBASE, Cochrane Library) were searched to identify studies published in the English language between May 2009- May 2019, which included patients (>18 years) with mRCC receiving AS of sunitinib. Data were analyzed using the random effect model and t-test. P < 0.05 was considered statistically significant. Results: Of 1922, 16 studies were included (eight AP, eight NAP). Among all grade AEs, mucositis (RR:0.22; 95% CI:0.12–0.40), cardiotoxicity (RR: 0.52; 95% CI: 0.31–0.88), nausea (RR:0.21; 95% CI: 0.10–0.44), hand-foot syndrome (RR:0.33; 95% CI:0.13–0.83), rash (RR: 0.52; 95% CI: 0.34–0.79), and aspartate transaminase (RR:0.57; 95% CI:0.33–0.98) were more common in AP. Leukopenia (RR:2.57; 95% CI:1.47–4.49), proteinemia (RR:4.45; 95% CI:2.12–9.33), and stomatitis (RR:4.33; 95% CI:2.6–7.23) occurred more commonly in NAP. Further, PFS was significantly longer in NAP, while longer OS was observed in AP (p < 0.001). Dose reduction was significantly higher in AP than NAP (52.08% vs. 40.6%, p = 0.0088). Conclusion: Safety profile of AS of sunitinib was similar with variations in the efficacy, dose reduction between AP and NAP. Sunitinib dose or schedule modification may mitigate AEs and enhance efficacy outcomes in mRCC by extending the treatment duration.
Collapse
Affiliation(s)
- Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Ishan Patel
- Medical Affairs, Oncology, Pfizer Emerging Asia, Mumbai, India
| | - Pratiksha Kapse
- Medical Affairs, Oncology, Pfizer Emerging Asia, Mumbai, India
| | - Manmohan Singh
- Regional Oncology Medical Lead, Pfizer Emerging Asia, Mumbai, India
| |
Collapse
|
12
|
Lv B, Chen J, Liu XL. Anlotinib-Induced Hypertension: Current Concepts and Future Prospects. Curr Pharm Des 2021; 28:216-224. [PMID: 34620054 DOI: 10.2174/1381612827666211006145141] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/27/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Anlotinib is a new tyrosine kinase inhibitor developed in China that targets the receptors for vascular endothelial growth factor, platelet-derived growth factor, fibroblast growth factor, and stem cell factor. Therefore, anlotinib inhibits tumor angiogenesis, representing a new therapeutic alternative for lung cancer. Hypertension is one of its most common adverse effects, leading to discontinuation of the drug and limited clinical usefulness. OBJECTIVE The present review aims to summarize the evidence on the prevalence, physiopathology, and management of anlotinib-induced hypertension, as well as its effect on the cancer prognosis. METHOD Searches in Medline, Cochrane Central Library, and Embase were performed using the following terms: anlotinib, adverse effect, hypertension, clinical trial, vascular endothelial growth factor, and antiangiogenic drugs. Citations were also identified by checking the reference sections of selected papers. RESULTS Except for a phase I clinical trial with a small sample size (n = 6), almost all the clinical trials on anlotinib have reported the development of anlotinib-induced hypertension. In these trials, the incidence of hypertension ranged from 13% to 67.7%, and that of grade 3/4 hypertension ranged from4.8% to 16%. Alterations in nitric oxide, endothelin-1, microvascular rarefaction, selective vasoconstrictions, and renal injury have been cited as potential mechanisms leading to anlotinib-induced hypertension. When needed, treatment may include general hygienic measures and pharmacotherapy in some cases. CONCLUSIONS To effectively manage anlotinib-induced hypertension, early prevention, a reasonable dosage regimen, and appropriate treatment are critical to effectively manage anlotinib-induced hypertension. Additionally, anlotinib-induced hypertension may be considered a marker for predicting efficacy.
Collapse
Affiliation(s)
- Bing Lv
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin Province. China
| | - Jing Chen
- Department of Endocrinology and Nephrology, Central Hospital of Tonghua, Tonghua, Jilin Province. China
| | - Xiao-Liang Liu
- Emergency Department, First Hospital of Jilin University, Changchun, Jilin Province. China
| |
Collapse
|
13
|
Sun F, Chen Z, Yao P, Weng B, Liu Z, Cheng L. Meta-Analysis of ABCG2 and ABCB1 Polymorphisms With Sunitinib-Induced Toxicity and Efficacy in Renal Cell Carcinoma. Front Pharmacol 2021; 12:641075. [PMID: 33762959 PMCID: PMC7982400 DOI: 10.3389/fphar.2021.641075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 01/19/2021] [Indexed: 11/13/2022] Open
Abstract
Background: ABCG2 and ABCB1 are genes related to the pharmacokinetics of sunitinib and have been associated with its toxicity and efficacy. However, the results have been controversial. This study aimed to evaluate the associations of ABCG2 and ABCB1 polymorphisms with sunitinib-induced toxicity and efficacy in renal cell carcinoma (RCC) by meta-analysis. Methods: PubMed, EMBASE, Cochrane Library, and Web of Science were systematically searched for studies investigating the associations of the ABCG2 rs2231142 polymorphism with sunitinib-induced toxicity and the associations of the ABCB1 rs1128503 and ABCB1 rs2032582 polymorphisms with sunitinib-induced toxicity and clinical outcomes. The associations were evaluated by effect size (ES) with 95% confidence intervals (CIs). Results: Eight and five studies were included in the toxicity and efficacy analysis, respectively, including a total of 1081 RCC patients. The ABCG2 rs2231142 A allele was associated with an increased risk of sunitinib-induced thrombocytopenia and hand-foot syndrome (HFS) in Asians (ES = 1.65, 95% CI = 1.15-2.36, p = 0.006; ES = 1.52, 95% CI = 1.02-2.27, p = 0.041). However, the ABCG2 rs2231142 polymorphism was not associated with sunitinib-induced hypertension or neutropenia (ES = 1.09, 95% CI = 0.69-1.73, p = 0.701; ES = 0.87, 95% CI = 0.57-1.31, p = 0.501). Compared with the C allele, the ABCB1 rs1128503 T allele was associated with a decreased risk of sunitinib-induced hypertension but worse progression-free survival (PFS) (ES = 0.44, 95% CI = 0.26-0.77, p = 0.004; ES = 1.36, 95% CI = 1.07-1.73, p = 0.011). There was no significant association between the T allele or C allele of ABCB1 rs1128503 and overall survival (OS) (ES = 0.82, 95% CI = 0.61-1.10, p = 0.184). The ABCB1 rs2032582 T allele was associated with worse PFS than the other alleles (ES = 1.46, 95% CI = 1.14-1.87, p = 0.003), while there was no significant association between the T allele or other alleles and sunitinib-induced hypertension, HFS, or OS (ES = 0.77, 95% CI = 0.46-1.29, p = 0.326; ES = 1.02, 95% CI = 0.65-1.62, p = 0.919; ES = 1.32, 95% CI = 0.85-2.05, p = 0.215). Conclusion: The results indicate that the ABCG2 rs2231142 polymorphism may serve as a predictor of sunitinib-induced thrombocytopenia and HFS in Asians, while the ABCB1 rs1128503 polymorphism may serve as a predictor of sunitinib-induced hypertension, and both the ABCB1 rs1128503 and rs2032582 polymorphisms may serve as predictors of PFS in RCC. These results suggest a possible application of individualized use of sunitinib according to the genetic background of patients.
Collapse
Affiliation(s)
- Fengjun Sun
- Department of Pharmacy, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhuo Chen
- Department of Pharmacy, Chongqing Emergency Medical Center, Chongqing, China
| | - Pu Yao
- Department of Pharmacy, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Bangbi Weng
- Department of Pharmacy, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhirui Liu
- Department of Pharmacy, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Lin Cheng
- Department of Pharmacy, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
14
|
Prevalence of pathogenic germline variants in patients with metastatic renal cell carcinoma. Genet Med 2021; 23:698-704. [DOI: 10.1038/s41436-020-01062-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 01/24/2023] Open
|
15
|
Visceral Adiposity as a Significant Predictor of Sunitinib-Induced Dose-Limiting Toxicities and Survival in Patients with Metastatic Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12123602. [PMID: 33276522 PMCID: PMC7761595 DOI: 10.3390/cancers12123602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022] Open
Abstract
Sunitinib is a first-line treatment for metastatic renal cell carcinoma (mRCC). Little is known about the predictive factors of sunitinib-induced dose-limiting toxicity (DLT) in Asian populations. We investigated whether body composition predicts sunitinib-induced DLT. We retrospectively reviewed sunitinib-treated Korean patients with clear cell mRCC from eight institutions. Body composition was measured using computed tomography. DLT was defined as any adverse event leading to dose reduction or treatment discontinuation. Univariate analysis was used to compare body composition indices, and logistic regression analyses were performed for factors predicting early DLT. Overall, 111/311 (32.5%) of patients experienced DLT. Significant differences were observed in the subcutaneous adipose tissue index (SATI; p = 0.001) and visceral adipose tissue index (VATI; p < 0.001) between patients with and without DLT. Multivariate analyses revealed that VATI (odds ratio: 1.013; p = 0.029) was significantly associated with early DLT. Additionally, 20% of patients who had a body mass index (BMI) greater than 23 kg/m2 and a low VATI experienced DLT, whereas 34.3% of the remaining groups had DLT (p = 0.034). Significant differences were observed for median progression-free survival (13.0 vs. 26.0 months, respectively; p = 0.006) between patients with low and high VATI. Visceral adiposity was a significant predictor of sunitinib-associated DLT and survival. Patients with a low VATI and a BMI greater than 23 kg/m2 experienced lower DLTs.
Collapse
|
16
|
Beuselinck B, Van Brussel T, Verbiest A, Vanmechelen M, Couchy G, Oudard S, Elaïdi R, Roussel E, Albersen M, Debruyne P, Baldewijns M, Machiels JP, Richard V, Verschaeve V, Wolter P, Rioux-Leclercq N, Laguerre B, Zucman-Rossi J, Lambrechts D. Validation of the Correlation Between Single Nucleotide Polymorphism rs307826 in VEGFR3 and Outcome in Metastatic Clear-Cell Renal Cell Carcinoma Patients Treated with Sunitinib. KIDNEY CANCER 2020. [DOI: 10.3233/kca-200086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Previously, we have shown a correlation between single nucleotide polymorphism (SNP) rs307826 in vascular endothelial growth factor receptor-3 (VEGFR3) and outcome in metastatic clear-cell renal cell carcinoma (m-ccRCC) patients treated with sunitinib. OBJECTIVE: We aimed to validate this finding in an independent patient series. METHODS: m-ccRCC patients receiving sunitinib as first-line targeted therapy were included in a validation cohort. Endpoints were response rate (RR), progression-free survival (PFS) and overall survival (OS). We also updated survival data of our discovery cohort as described previously. RESULTS: Eighty-four patients were included in the validation cohort. rs307826 AG/GG-carriers had a shorter PFS (8 versus 12 months, p = 0.04) and a trend towards a shorter OS (18 versus 27 months, p = 0.22) compared to AA-carriers. In the total series of 168 patients (from the discovery cohort, as described previously, and the validation cohort), rs307826 AG/GG-carriers had a poorer RR (29% versus 53%, p = 0.008), PFS (8 versus 15 months, p = 0.0002) and OS (22 versus 31 months, p = 0.004) compared to AA-carriers. rs307826 was independently associated with PFS and OS on multivariate analysis. CONCLUSION: VEGFR3 rs307826 seems to be associated with outcome on sunitinib in m-ccRCC. Its impact highlights the role of VEGFR3 in ccRCC pathogenesis and as a target of sunitinib.
Collapse
Affiliation(s)
- Benoit Beuselinck
- Department of General Medical Oncology and Laboratory for Experimental Oncology, University Hospitals Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Thomas Van Brussel
- Department of Oncology, Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium
- Vesalius Research Center, VIB, Leuven, Belgium
| | - Annelies Verbiest
- Department of General Medical Oncology and Laboratory for Experimental Oncology, University Hospitals Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Maxime Vanmechelen
- Department of General Medical Oncology and Laboratory for Experimental Oncology, University Hospitals Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | | - Stéphane Oudard
- Department of Medical Oncology, Georges Pompidou European Hospital, Université Paris-5 René Descartes, Paris, France
| | - Reza Elaïdi
- Department of Medical Oncology, Georges Pompidou European Hospital, Université Paris-5 René Descartes, Paris, France
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Philip Debruyne
- Department of Medical Oncology, AZ Groeninge, Kortrijk, Belgium, and Faculty of Health, Education, Medicine & Social Care, Anglia Ruskin University, Chelmsford, UK
| | - Marcella Baldewijns
- Department of Pathology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Jean-Pascal Machiels
- Department of Medical Oncology, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Vincent Richard
- Department of Medical Oncology, CHU Ambroise Paré, Mons, Belgium
| | - Vincent Verschaeve
- Department of Medical Oncology, Grand Hôpital de Charleroi, Charleroi, Belgium
| | - Pascal Wolter
- Department of Medical Oncology, St. Nikolaus-Hospital Eupen, Eupen, Belgium
| | | | | | | | - Diether Lambrechts
- Department of Oncology, Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium
- Vesalius Research Center, VIB, Leuven, Belgium
| |
Collapse
|
17
|
Hopkins AM, Menz BD, Wiese MD, Kichenadasse G, Gurney H, McKinnon RA, Rowland A, Sorich MJ. Nuances to precision dosing strategies of targeted cancer medicines. Pharmacol Res Perspect 2020; 8:e00625. [PMID: 32662214 PMCID: PMC7358594 DOI: 10.1002/prp2.625] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 01/05/2023] Open
Abstract
Selecting the dose of a targeted cancer medicine that is most appropriate for a specific individual is a rational approach to maximize therapeutic outcomes and minimize toxicity. There are many different options for optimizing the dose of targeted cancer medicines and the purpose of this review is to provide a comprehensive comparison of the main options explored in prospective studies. Precision initial dose selection of targeted cancer therapies has been minimally explored to date; however, concentration, toxicity, and therapeutic outcome markers are used to guide on-therapy dose adaption of targeted cancer therapies across several medicines and cancers. While a specific concentration, toxicity, or therapeutic outcome marker commonly dominates an investigated precision on-therapy dose adaption strategy, greater attention to simultaneously account for exposure, toxicity, therapeutic outcomes, disease status, time since treatment initiation and patient preferences are required for optimal patient outcomes. To enable successful implementation of precision dosing strategies for targeted cancer medicines into clinical practice, future prospective studies aiming to develop strategies should consider these elements in their design.
Collapse
Affiliation(s)
- Ashley M. Hopkins
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Bradley D. Menz
- Division of PharmacySouthern Adelaide Local Health Network, Flinders Medical CentreAdelaideSouth AustraliaAustralia
| | - Michael D. Wiese
- School of Pharmacy and Medical SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Ganessan Kichenadasse
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Howard Gurney
- Department of Medical OncologyWestmead HospitalSydneyNew South WalesAustralia
| | - Ross A. McKinnon
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Michael J. Sorich
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| |
Collapse
|
18
|
Anti-angiogenesis and Immunotherapy: Novel Paradigms to Envision Tailored Approaches in Renal Cell-Carcinoma. J Clin Med 2020; 9:jcm9051594. [PMID: 32456352 PMCID: PMC7291047 DOI: 10.3390/jcm9051594] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022] Open
Abstract
Although decision making strategy based on clinico-histopathological criteria is well established, renal cell carcinoma (RCC) represents a spectrum of biological ecosystems characterized by distinct genetic and molecular alterations, diverse clinical courses and potential specific therapeutic vulnerabilities. Given the plethora of drugs available, the subtype-tailored treatment to RCC subtype holds the potential to improve patient outcome, shrinking treatment-related morbidity and cost. The emerging knowledge of the molecular taxonomy of RCC is evolving, whilst the antiangiogenic and immunotherapy landscape maintains and reinforces their potential. Although several prognostic factors of survival in patients with RCC have been described, no reliable predictive biomarkers of treatment individual sensitivity or resistance have been identified. In this review, we summarize the available evidence able to prompt more precise and individualized patient selection in well-designed clinical trials, covering the unmet need of medical choices in the era of next-generation anti-angiogenesis and immunotherapy.
Collapse
|
19
|
Garrigós C, Molina-Pinelo S, Meléndez R, Espinosa M, Lerma A, Taron M, García-Donas J, Rodriguez-Antona C, Duran I. MicroRNAs as potential predictors of extreme response to tyrosine kinase inhibitors in renal cell cancer. Urol Oncol 2020; 38:640.e23-640.e29. [PMID: 32081561 DOI: 10.1016/j.urolonc.2020.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/21/2020] [Accepted: 01/25/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND MicroRNAs play an important role as modulators of gene expression in several biological processes and are closely related to development and cell differentiation regulation. Previous works have revealed a potential predictive role for miRNAs in different tumor types. This study aims to analyze the ability of miRNAs in segregating metastatic renal cell carcinoma patients according to their responses to tyrosine kinase inhibitors (TKIs). METHODS Extreme responders were considered in the study and were defined as those patients that either had a long-term response (LR) (progression-free survival ˃11 months) or those that were primary refractory (PR) (progression as best response). The expression of 754 miRNAs was analyzed in tumor tissue of these 2 sets of patients. RESULTS In a study cohort (n = 15) 4 miRNAs were significantly associated with patient response and differentially expressed in PR vs. LR (up-regulated in PR vs. LR: miR-425-5p, down-regulated in PR vs. LR: miR-139-3p, let-7d and let-7e). Further analysis in a validation cohort (n = 36) revealed similar results. CONCLUSION The present data strength the potential role of miRNAs as a tool to predict treatment outcomes in patients with metastatic renal cell carcinoma treated with TKIs.
Collapse
Affiliation(s)
- Carmen Garrigós
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Sonia Molina-Pinelo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Ricardo Meléndez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Marta Espinosa
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Antonio Lerma
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Miguel Taron
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Jesús García-Donas
- Department of Medical Oncology, HM Hospitales - Centro Integral Oncológico HM Clara Campal, Madrid, Spain
| | - Cristina Rodriguez-Antona
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain; ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Ignacio Duran
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain.
| |
Collapse
|
20
|
Westerdijk K, Desar IME, Steeghs N, van der Graaf WTA, van Erp NP. Imatinib, sunitinib and pazopanib: From flat-fixed dosing towards a pharmacokinetically guided personalized dose. Br J Clin Pharmacol 2020; 86:258-273. [PMID: 31782166 PMCID: PMC7015742 DOI: 10.1111/bcp.14185] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/21/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are anti‐cancer drugs that target tyrosine kinases, enzymes that are involved in multiple cellular processes. Currently, multiple oral TKIs have been introduced in the treatment of solid tumours, all administered in a fixed dose, although large interpatient pharmacokinetic (PK) variability is described. For imatinib, sunitinib and pazopanib exposure‐treatment outcome (efficacy and toxicity) relationships have been established and therapeutic windows have been defined, therefore dose optimization based on the measured blood concentration, called therapeutic drug monitoring (TDM), can be valuable in increasing efficacy and reducing the toxicity of these drugs. In this review, an overview of the current knowledge on TDM guided individualized dosing of imatinib, sunitinib and pazopanib for the treatment of solid tumours is presented. We summarize preclinical and clinical data that have defined thresholds for efficacy and toxicity. Furthermore, PK models and factors that influence the PK of these drugs which partly explain the interpatient PK variability are summarized. Finally, pharmacological interventions that have been performed to optimize plasma concentrations are described. Based on current literature, we advise which methods should be used to optimize exposure to imatinib, sunitinib and pazopanib.
Collapse
Affiliation(s)
- Kim Westerdijk
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek hospital, Amsterdam, the Netherlands
| | - Winette T A van der Graaf
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Medical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek hospital, Amsterdam, the Netherlands
| | - Nielka P van Erp
- Department of Clinical Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | | |
Collapse
|
21
|
Grépin R, Guyot M, Dumond A, Durivault J, Ambrosetti D, Roussel JF, Dupré F, Quintens H, Pagès G. The combination of bevacizumab/Avastin and erlotinib/Tarceva is relevant for the treatment of metastatic renal cell carcinoma: the role of a synonymous mutation of the EGFR receptor. Theranostics 2020; 10:1107-1121. [PMID: 31938054 PMCID: PMC6956821 DOI: 10.7150/thno.38346] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 08/14/2019] [Indexed: 01/25/2023] Open
Abstract
Metastatic clear cell renal cell carcinomas (mRCC) over-express the vascular endothelial growth factor (VEGF). Hence, the anti-VEGF antibody bevacizumab/Avastin (BVZ) combined with interferon alpha (IFN) was approved for the treatment of mRCC. However, approval was lost in July 2016 due to the absence of sustained efficacy. We previously showed that BVZ accelerates tumor growth in experimental models of mRCC in mice, results in part explained by down-regulation of the phospho tyrosine phosphatase receptor kappa (PTPRκ) in tumor cells. The epidermal growth factor receptor (EGFR) is a direct target of PTPRκ. Its down-regulation leads to constitutive activation of EGFR, an observation which prompted us to test the effect of the EGFR inhibitor erlotinib/Tarceva (ERLO) in addition to BVZ/IFN. The influence of the long non-coding RNA, EGFR-AS1, on ERLO efficacy was also addressed. Methods: The effect of BVZ/IFN/ERLO was tested on the growth of experimental tumors in nude mice. The presence of germline mutation in the EGFR was evaluated on cell lines and primary RCC cells. In vitro translation and transfections of expression vectors coding the wild-type or the EGFR mutated gene in HEK-293 cells were used to test the role of EGFR mutation of the ERLO efficacy. Correlation between EGFR/EGFR-AS1 expression and survival was analyzed with an online available data base (TCGA). Results: Tumor growth was strongly reduced by the triple combination BVZ/IFN/ERLO and linked to reduced levels of pro-angiogenic/pro-inflammatory cytokines of the ELR+CXCL family and to subsequent inhibition of vascularization, a decreased number of lymphatic vessels and polarization of macrophages towards the M1 phenotype. Cells isolated from surgical resection of human tumors presented a range of sensitivity to ERLO depending on the presence of a newly detected mutation in the EGFR and to the presence of EGFR-AS1. Conclusions: Our results point-out that the BVZ/IFN/ERLO combination deserves testing for the treatment of mRCC that have a specific mutation in the EGFR.
Collapse
|
22
|
D'Aniello C, Berretta M, Cavaliere C, Rossetti S, Facchini BA, Iovane G, Mollo G, Capasso M, Pepa CD, Pesce L, D'Errico D, Buonerba C, Di Lorenzo G, Pisconti S, De Vita F, Facchini G. Biomarkers of Prognosis and Efficacy of Anti-angiogenic Therapy in Metastatic Clear Cell Renal Cancer. Front Oncol 2019; 9:1400. [PMID: 31921657 PMCID: PMC6917607 DOI: 10.3389/fonc.2019.01400] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/27/2019] [Indexed: 12/30/2022] Open
Abstract
In the last decades, the prognosis of metastatic renal cell carcinoma (mRCC) has remarkably improved following the advent of the "targeted therapy" era. The expanding knowledge on the prominent role played by angiogenesis in RCC pathogenesis has led to approval of multiple anti-angiogenic agents such as sunitinib, pazopanib, axitinib, cabozantinib, sorafenib, and bevacizumab. These agents can induce radiological responses and delay cancer progression for months or years before onset of resistance, with a clinically meaningful activity. The need for markers of prognosis and efficacy of anti-angiogenic agents has become more compelling as novel systemic immunotherapy agents have also been approved in RCC and can be administered as an alternative to angiogenesis inhibitors. Anti PD-1 monoclonal antibody nivolumab has been approved in the second-line setting after tyrosine kinase inhibitors failure, while combination of nivolumab plus anti CTLA-4 monoclonal antibody ipilimumab has been approved as first-line therapy of RCC patients at intermediate or poor prognosis. In this review article, biomarkers of prognosis and efficacy of antiangiogenic therapies are summarized with a focus on those that have the potential to affect treatment decision-making in RCC. Biomarkers predictive of toxicity of anti-angiogenic agents have also been discussed.
Collapse
Affiliation(s)
- Carmine D'Aniello
- Division of Medical Oncology, A.O.R.N. dei COLLI “Ospedali Monaldi-Cotugno-CTO,”Naples, Italy
| | - Massimiliano Berretta
- Division of Medical Oncology, Istituto Nazionale Tumori, IRCCS CRO Aviano (PN), Milan, Italy
| | - Carla Cavaliere
- UOC of Medical Oncology, ASL NA 3 SUD, Ospedali Riuniti Area Nolana, Nola, Italy
| | - Sabrina Rossetti
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Bianca Arianna Facchini
- Division of Medical Oncology, Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Gelsomina Iovane
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Giovanna Mollo
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Mariagrazia Capasso
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | | | - Laura Pesce
- Oncology Unit, San Luca Hospital, Vallo Della Lucania, Italy
| | - Davide D'Errico
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Carlo Buonerba
- CRTR Rare Tumors Reference Center, AOU Federico II, Naples, Italy
- Environment & Health Operational Unit, Zoo-Prophylactic Institute of Southern Italy, Portici, Italy
| | - Giuseppe Di Lorenzo
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
- Department of Medicine, University of Molise, Campobasso, Italy
| | - Salvatore Pisconti
- Department of Onco-Hematology, Medical Oncology, S.G. Moscati Hospital, Taranto, Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Gaetano Facchini
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| |
Collapse
|
23
|
Buller DM, Ristau BT. Improving patient selection for adjuvant therapy in high-risk renal cell carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S104. [PMID: 31576311 DOI: 10.21037/atm.2019.04.75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Dylan M Buller
- University of Connecticut School of Medicine, Farmington, CT, USA
| | | |
Collapse
|
24
|
Werbrouck E, Bastin J, Lambrechts D, Verbiest A, Van Brussel T, Lerut E, Machiels JP, Verschaeve V, Richard V, Debruyne PR, Decallonne B, Schöffski P, Bechter O, Wolter P, Beuselinck B. ABCG2 Polymorphism rs2231142 and hypothyroidism in metastatic renal cell carcinoma patients treated with sunitinib. Acta Clin Belg 2019; 74:180-188. [PMID: 29792121 DOI: 10.1080/17843286.2018.1477229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIM Vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) cause significant adverse events including thyroid dysfunction, mainly hypothyroidism, in a considerable proportion of patients. In a series of metastatic renal cell carcinoma (mRCC) patients treated with sunitinib, we aimed to study the correlation between hypothyroidism and single nucleotide polymorphisms (SNPs) in genes involved in sunitinib pharmacokinetics and pharmacodynamics. PATIENTS AND METHODS We included 79 mRCC patients who started sunitinib between November 2005 and March 2016. Serum thyroid function markers were collected at start and during sunitinib therapy. Germ-line DNA genotyping for 16 SNPs in 8 candidate genes was performed. Endpoints were time to increase in thyroid stimulating hormone (TSH) and time to decrease in T4 or free T4 (FT4) on day 1 and day 28 of each sunitinib cycle. RESULTS Patients with the ABCG2 rs2231142 CC-genotype had a significantly longer time-to-TSH-increase on day 1 (11 vs. 5 cycles; p = 0.0011), and time-to-T4/FT4-decrease on day 1 (not reached vs. 10 cycles; p = 0.013) and day 28 (28 vs. 7 cycles; p = 0.03) compared to CA-carriers. Patients with the CYP3A5 rs776746 GG-genotype had a significantly longer time-to-TSH-increase at day 1 compared to GA-patients: 11 vs. 5 cycles (p = 0.0071). Significant associations were also found between PDGFRA rs35597368 and rs1800812 and time-to-TSH-increase at day 28. CONCLUSION Polymorphism rs2231142 in the efflux pump ABCG2 is associated with hypothyroidism in mRCC patients treated with sunitinib.
Collapse
Affiliation(s)
- Emilie Werbrouck
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute and Department of Oncology, KU Leuven, Leuven, Belgium
| | - Julie Bastin
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute and Department of Oncology, KU Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Oncology, KU Leuven, Leuven, Belgium
- Research Center, VIB, Leuven, Belgium
| | - Annelies Verbiest
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute and Department of Oncology, KU Leuven, Leuven, Belgium
| | - Thomas Van Brussel
- Laboratory for Translational Genetics, Department of Oncology, KU Leuven, Leuven, Belgium
- Research Center, VIB, Leuven, Belgium
| | - Evelyne Lerut
- Department of Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Jean-Pascal Machiels
- Department of Medical Oncology and Hematology, UCL Brussels and Hospitals Saint-Luc, Brussels, Belgium
| | | | - Vincent Richard
- Department of Medical Oncology, CHU Ambroise Paré, Mons, Belgium
| | - Philip R. Debruyne
- Department of Medical Oncology, General Hospital Groeninge, Kortrijk, Belgium
- Faculty of Health, Social Care & Education, Anglia Ruskin University, Chelmsford, UK
| | | | - Patrick Schöffski
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute and Department of Oncology, KU Leuven, Leuven, Belgium
| | - Oliver Bechter
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute and Department of Oncology, KU Leuven, Leuven, Belgium
| | - Pascal Wolter
- Department of Medical Oncology, Centre Hospitalier Regional Verviers East Belgium, Verviers, Belgium
| | - Benoit Beuselinck
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute and Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Fazio N, Martini JF, Croitoru AE, Schenker M, Li S, Rosbrook B, Fernandez K, Tomasek J, Thiis-Evensen E, Kulke M, Raymond E. Pharmacogenomic analyses of sunitinib in patients with pancreatic neuroendocrine tumors. Future Oncol 2019; 15:1997-2007. [DOI: 10.2217/fon-2018-0934] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: Evaluate associations between clinical outcomes and SNPs in patients with well-differentiated pancreatic neuroendocrine tumors receiving sunitinib. Patients & methods: Kaplan–Meier and Cox proportional hazards models were used to analyze the association between SNPs and survival outcomes using data from a sunitinib Phase IV (genotyped, n = 56) study. Fisher’s exact test was used to analyze objective response rate and genotype associations. Results: After multiplicity adjustment, progression-free and overall survivals were not significantly correlated with SNPs; however, a higher objective response rate was significantly associated with IL1B rs16944 G/A versus G/G (46.4 vs 4.5%; p = 0.001). Conclusion: IL1B SNPs may predict treatment response in patients with pancreatic neuroendocrine tumors. VEGF pathway SNPs are potentially associated with survival outcomes.
Collapse
Affiliation(s)
- Nicola Fazio
- Division of Gastrointestinal Medical Oncology & Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | | | - Adina E Croitoru
- Department of Medical Oncology, Fundeni Clinical Institute, Bucharest, Romania
| | - Michael Schenker
- Centrul de Oncologie Sf. Nectarie, Oncologie Medicala, Craiova, Romania
| | | | | | | | - Jiri Tomasek
- Faculty of Medicine, Masaryk Memorial Cancer Institute, Masaryk University, Brno, Czech Republic
| | - Espen Thiis-Evensen
- Department of Gastroenterology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Matthew Kulke
- Boston University & Boston Medical Center, Boston, MA, USA
| | - Eric Raymond
- Department of Medical Oncology, Paris Saint-Joseph Hospital Group, Paris, France
| |
Collapse
|
26
|
Vanmechelen M, Lambrechts D, Van Brussel T, Verbiest A, Couchy G, Schöffski P, Dumez H, Debruyne PR, Lerut E, Machiels JP, Richard V, Albersen M, Verschaeve V, Oudard S, Méjean A, Wolter P, Zucman-Rossi J, Beuselinck B. Fibroblast Growth Factor Receptor-2 Polymorphism rs2981582 is Correlated With Progression-free Survival and Overall Survival in Patients With Metastatic Clear-cell Renal Cell Carcinoma Treated With Sunitinib. Clin Genitourin Cancer 2019; 17:e235-e246. [DOI: 10.1016/j.clgc.2018.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023]
|
27
|
Wei JH, Feng ZH, Cao Y, Zhao HW, Chen ZH, Liao B, Wang Q, Han H, Zhang J, Xu YZ, Li B, Wu JT, Qu GM, Wang GP, Liu C, Xue W, Liu Q, Lu J, Li CX, Li PX, Zhang ZL, Yao HH, Pan YH, Chen WF, Xie D, Shi L, Gao ZL, Huang YR, Zhou FJ, Wang SG, Liu ZP, Chen W, Luo JH. Predictive value of single-nucleotide polymorphism signature for recurrence in localised renal cell carcinoma: a retrospective analysis and multicentre validation study. Lancet Oncol 2019; 20:591-600. [PMID: 30880070 DOI: 10.1016/s1470-2045(18)30932-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/26/2018] [Accepted: 12/10/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Identification of high-risk localised renal cell carcinoma is key for the selection of patients for adjuvant treatment who are at truly higher risk of reccurrence. We developed a classifier based on single-nucleotide polymorphisms (SNPs) to improve the predictive accuracy for renal cell carcinoma recurrence and investigated whether intratumour heterogeneity affected the precision of the classifier. METHODS In this retrospective analysis and multicentre validation study, we used paraffin-embedded specimens from the training set of 227 patients from Sun Yat-sen University (Guangzhou, Guangdong, China) with localised clear cell renal cell carcinoma to examine 44 potential recurrence-associated SNPs, which were identified by exploratory bioinformatics analyses of a genome-wide association study from The Cancer Genome Atlas (TCGA) Kidney Renal Clear Cell Carcinoma (KIRC) dataset (n=114, 906 600 SNPs). We developed a six-SNP-based classifier by use of LASSO Cox regression, based on the association between SNP status and patients' recurrence-free survival. Intratumour heterogeneity was investigated from two other regions within the same tumours in the training set. The six-SNP-based classifier was validated in the internal testing set (n=226), the independent validation set (Chinese multicentre study; 428 patients treated between Jan 1, 2004 and Dec 31, 2012, at three hospitals in China), and TCGA set (441 retrospectively identified patients who underwent resection between 1998 and 2010 for localised clear cell renal cell carcinoma in the USA). The main outcome was recurrence-free survival; the secondary outcome was overall survival. FINDINGS Although intratumour heterogeneity was found in 48 (23%) of 206 cases in the internal testing set with complete SNP information, the predictive accuracy of the six-SNP-based classifier was similar in the three different regions of the training set (areas under the curve [AUC] at 5 years: 0·749 [95% CI 0·660-0·826] in region 1, 0·734 [0·651-0·814] in region 2, and 0·736 [0·649-0·824] in region 3). The six-SNP-based classifier precisely predicted recurrence-free survival of patients in three validation sets (hazard ratio [HR] 5·32 [95% CI 2·81-10·07] in the internal testing set, 5·39 [3·38-8·59] in the independent validation set, and 4·62 [2·48-8·61] in the TCGA set; all p<0·0001), independently of patient age or sex and tumour stage, grade, or necrosis. The classifier and the clinicopathological risk factors (tumour stage, grade, and necrosis) were combined to construct a nomogram, which had a predictive accuracy significantly higher than that of each variable alone (AUC at 5 years 0·811 [95% CI 0·756-0·861]). INTERPRETATION Our six-SNP-based classifier could be a practical and reliable predictor that can complement the existing staging system for prediction of localised renal cell carcinoma recurrence after surgery, which might enable physicians to make more informed treatment decisions about adjuvant therapy. Intratumour heterogeneity does not seem to hamper the accuracy of the six-SNP-based classifier as a reliable predictor of recurrence. The classifier has the potential to guide treatment decisions for patients at differing risks of recurrence. FUNDING National Key Research and Development Program of China, National Natural Science Foundation of China, Guangdong Provincial Science and Technology Foundation of China, and Guangzhou Science and Technology Foundation of China.
Collapse
Affiliation(s)
- Jin-Huan Wei
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zi-Hao Feng
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yun Cao
- Department of Pathology, Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hong-Wei Zhao
- Department of Urology, Affiliated Yantai Yuhuangding Hospital, Qingdao University Medical College, Shandong, China
| | - Zhen-Hua Chen
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Bing Liao
- Department of Pathology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qing Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Hui Han
- Department of Urology, Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jin Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yun-Ze Xu
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ji-Tao Wu
- Department of Urology, Affiliated Yantai Yuhuangding Hospital, Qingdao University Medical College, Shandong, China
| | - Gui-Mei Qu
- Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University Medical College, Shandong, China
| | - Guo-Ping Wang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Cong Liu
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Lu
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cai-Xia Li
- School of Mathematics and Computational Science, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Pei-Xing Li
- School of Mathematics and Computational Science, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhi-Ling Zhang
- Department of Urology, Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hao-Hua Yao
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yi-Hui Pan
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wen-Fang Chen
- Department of Pathology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dan Xie
- Department of Pathology, Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lei Shi
- Department of Urology, Affiliated Yantai Yuhuangding Hospital, Qingdao University Medical College, Shandong, China
| | - Zhen-Li Gao
- Department of Urology, Affiliated Yantai Yuhuangding Hospital, Qingdao University Medical College, Shandong, China
| | - Yi-Ran Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fang-Jian Zhou
- Department of Urology, Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shao-Gang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Zhi-Ping Liu
- Department of Internal Medicine and Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Wei Chen
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun-Hang Luo
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
28
|
Kammerer-Jacquet SF, Brunot A, Lefort M, Bayat S, Peyronnet B, Verhoest G, Mathieu R, Lespagnol A, Mosser J, Laguerre B, Ravaud A, Bernhard JC, Dupuis F, Yacoub M, Belaud-Rotureau MA, Bensalah K, Rioux-Leclercq N. Metastatic Clear-cell Renal Cell Carcinoma With a Long-term Response to Sunitinib: A Distinct Phenotype Independently Associated With Low PD-L1 Expression. Clin Genitourin Cancer 2019; 17:169-176.e1. [PMID: 30837208 DOI: 10.1016/j.clgc.2019.01.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 01/26/2019] [Accepted: 01/26/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Long-term responders (LTRs) are defined by at least 18 months of response to sunitinib in metastatic clear-cell renal cell carcinoma (ccRCC). Well-described by clinical studies, the phenotype of these tumors has never been explored. PATIENTS AND METHODS In a retrospective and multicenter study, 90 ccRCCs of patients with metastatic disease were analyzed. Immunohistochemistry (carbonic anhydrase IX, vascular endothelial growth factor, c-MET, programmed death-ligand 1 [PD-L1], and PD-1) and VHL status were performed. Progression-free survival and overall survival were calculated from sunitinib introduction and from progression. LTRs and their corresponding tumors were compared with others using univariate and multivariate analysis. RESULTS Twenty-eight patients were LTRs. They had a median progression-free survival of 28 months versus 4 months for other patients (P < .001). Similarly, LTRs had a median overall survival of 49 months versus 14 months (P < .001), even from progression (median, 21 vs. 7 months; P = .029). They were associated with a favorable or intermediate risk (International Metastatic Renal Cell Carcinoma Database Consortium model) (P = .007) and less liver metastasis (P = .036). They experienced more frequent complete or partial responses at the first radiologic evaluation (P = .035). The corresponding ccRCCs were associated with less nucleolar International Society for Urological Pathology grade 4 (P = .037) and hilar fat infiltration (P = .006). They were also associated with low PD-L1 expression (P = .02). Only the International Metastatic Renal Cell Carcinoma Database Consortium model and PD-L1 expression remained significant after multivariate analysis (P = .014 and P = .029, respectively). CONCLUSION Primary tumor characteristics of LTRs were studied for the first time and demonstrated a different phenotype. Interestingly, they were characterized by low expression of PD-L1, suggesting a potentially lower impact of targeted immunotherapy in these patients.
Collapse
Affiliation(s)
- Solène-Florence Kammerer-Jacquet
- Service d'Anatomie et Cytologie Pathologiques, Université de Rennes 1, Université Bretagne Loire, Rennes, France; Unité Mixte de Recherche 6290-Institut de Génétique et Développement de Rennes, Rennes, France.
| | - Angelique Brunot
- Service d'Oncologie Médicale, Centre Eugène Marquis, Rennes, France
| | | | - Sahar Bayat
- Ecole des Hautes Etudes en Santé Publique, Rennes, France
| | - Benoit Peyronnet
- Service d'Urologie, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Gregory Verhoest
- Service d'Urologie, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Romain Mathieu
- Service d'Urologie, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Alexandra Lespagnol
- Service de Génétique Somatique des Cancers, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Jean Mosser
- Service de Génétique Somatique des Cancers, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | | | - Alain Ravaud
- Service d'Oncologie Médicale, Centre Hospitalier Universitaire Saint-André, Bordeaux, France
| | | | - Frantz Dupuis
- Service d'Anatomie et Cytologie Pathologiques, Centre Hospitalier Universitaire Pellegrin, Bordeaux, France
| | - Mokrane Yacoub
- Service d'Anatomie et Cytologie Pathologiques, Centre Hospitalier Universitaire Pellegrin, Bordeaux, France
| | - Marc-Antoine Belaud-Rotureau
- Unité Mixte de Recherche 6290-Institut de Génétique et Développement de Rennes, Rennes, France; Service de Cytogénétique, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Karim Bensalah
- Service d'Urologie, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Nathalie Rioux-Leclercq
- Service d'Anatomie et Cytologie Pathologiques, Université de Rennes 1, Université Bretagne Loire, Rennes, France; Unité Mixte de Recherche 6290-Institut de Génétique et Développement de Rennes, Rennes, France
| |
Collapse
|
29
|
Falkowski S. Résistance aux inhibiteurs des tyrosines kinases dans le cancer du rein. Bull Cancer 2019; 105 Suppl 3:S255-S260. [PMID: 30595154 DOI: 10.1016/s0007-4551(18)30380-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
FOCUS ON RESISTANCE TO TYROSINE KINASE INHIBITORS IN RENAL CANCER: The last decade has seen significant advances in understanding the biology and genetics of kidney cancer, some of which have radically changed treatment standards, including the emergence of targeted therapies. TKIs have significantly improved outcome in patients with metastatic disease. Nevertheless, a subset of patients (approximately 25 %) does not show any clinical benefit from targeted therapies. In many cases, patients initially respond to therapy but resistance to targeted agents has been shown to develop after a median of 6-12 months of treatment. Two general models of tumor resistance to anti-angiogenic agents targeting the VEGF pathway have been proposed: an adaptive (evasive) resistance, which occurs after a prolonged application of a drug (providing a period of tumor control), or intrinsic one (preexisting) non-responsiveness despite the presence of an active agent, showing no therapeutic benefit. Intrinsic resistance is related to tumor redundancy of pro-angiogenic pathways. Acquired resistance is associated with activation of alternative pathways either by upregulation of the VEGF pathway or by recruitment of alternative angiogenic factors responsible for tumor revascularization. Because different combinations and sequences of TKI are tested in clinical trials and immunotherapy (alone or in combination) radically alters patient management in its metastatic disease, the current effort aims at identifying resistance processes, evaluating their importance and proposing rational therapeutic approaches in order to obtain an additional clinical benefit. Our article summarizes the different mechanisms of resistance described in the literature.
Collapse
Affiliation(s)
- Sabrina Falkowski
- Service oncologie, polyclinique de Limoges, site Chénieux, 18, rue du Général-Catroux, 87000 Limoges, France.
| |
Collapse
|
30
|
Crona DJ, Skol AD, Leppänen VM, Glubb DM, Etheridge AS, Hilliard E, Peña CE, Peterson YK, Klauber-DeMore N, Alitalo KK, Innocenti F. Genetic Variants of VEGFA and FLT4 Are Determinants of Survival in Renal Cell Carcinoma Patients Treated with Sorafenib. Cancer Res 2019; 79:231-241. [PMID: 30385613 PMCID: PMC6541205 DOI: 10.1158/0008-5472.can-18-1089] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/03/2018] [Accepted: 10/29/2018] [Indexed: 12/30/2022]
Abstract
Molecular markers of sorafenib efficacy in patients with metastatic renal cell carcinoma (mRCC) are not available. The purpose of this study was to discover genetic markers of survival in patients with mRCC treated with sorafenib. Germline variants from 56 genes were genotyped in 295 patients with mRCC. Variant-overall survival (OS) associations were tested in multivariate regression models. Mechanistic studies were conducted to validate clinical associations. VEGFA rs1885657, ITGAV rs3816375, and WWOX rs8047917 (sorafenib arm), and FLT4 rs307826 and VEGFA rs3024987 (sorafenib and placebo arms combined) were associated with shorter OS. FLT4 rs307826 increased VEGFR-3 phosphorylation, membrane trafficking, and receptor activation. VEGFA rs1885657 and rs58159269 increased transcriptional activity of the constructs containing these variants in endothelial and RCC cell lines, and VEGFA rs58159269 increased endothelial cell proliferation and tube formation. FLT4 rs307826 and VEGFA rs58159269 led to reduced sorafenib cytotoxicity. Genetic variation in VEGFA and FLT4 could affect survival in sorafenib-treated patients with mRCC. These markers should be examined in additional malignancies treated with sorafenib and in other angiogenesis inhibitors used in mRCC. SIGNIFICANCE: Clinical and mechanistic data identify germline genetic variants in VEGFA and FLT4 as markers of survival in patients with metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Daniel J Crona
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, The University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Andrew D Skol
- The University of Chicago, Department of Medicine, Chicago, Illinois
| | | | - Dylan M Glubb
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, The University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina
- The Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Amy S Etheridge
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, The University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina
| | - Eleanor Hilliard
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Carol E Peña
- Bayer HealthCare Pharmaceuticals, Montville, New Jersey
| | - Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Nancy Klauber-DeMore
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Kari K Alitalo
- Wihuri Research Institute and University of Helsinki, Helsinki, Finland
| | - Federico Innocenti
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, The University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina.
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
31
|
Roles of pharmacogenomics in non-anthracycline antineoplastic-induced cardiovascular toxicities: A systematic review and meta-analysis of genotypes effect. Int J Cardiol 2018; 280:190-197. [PMID: 30594345 DOI: 10.1016/j.ijcard.2018.12.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 12/02/2018] [Accepted: 12/17/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Exploration on genetic roles in antineoplastic-related cardiovascular toxicity has increased with the advancement of genotyping technology. However, knowledge on the extent of genetic determinants in affecting the susceptibility to the cardiovascular toxicities of antineoplastic is limited. This study aims to identify potential single nucleotide polymorphism (SNP) in predicting non-anthracycline antineoplastic-related cardiovascular toxicity. METHODS We systematically searched for original research in PubMed, Cochrane Central Register of Controlled Studies, CINAHL Plus, EMBASE and HuGE Navigator from database inception until January 2018. Studies on association between polymorphism and antineoplastic-induced cardiovascular toxicity in patients treated for cancer of all antineoplastic agents were included except for anthracycline. Case reports, conference abstracts, reviews and non-patient studies were excluded. Data extracted by two independent reviewers were combined with random-effects model and reported according to PRISMA and MOOSE guidelines. RESULTS The 35 studies included examined a total of 219 SNPs in 80 genes, 11 antineoplastic and 5 types of cardiovascular toxicities. Meta-analyses showed that human epidermal growth factor receptor 2 (HER2) rs1136201, a risk variants (pooled OR: 2.43; 1.17-5.06, p = 0.018) is a potential predictors for trastuzumab-related cardiotoxicity. Gene dose effect analysis showed number of variant allele may contribute to the risk too. CONCLUSIONS This review found that HER2 rs1136201 can have the potential in predicting trastuzumab-related heart failure. As such, further studies are needed to confirm the validity of these results as well as determine the economic aspect of using SNPs prior to its implementation as a clinical practice.
Collapse
|
32
|
Jiménez-Fonseca P, Martín MN, Carmona-Bayonas A, Calvo A, Fernández-Mateos J, Redrado M, Capdevila J, Lago NM, Lacasta A, Muñarriz J, Segura Á, Fuster J, Barón F, Llanos M, Serrano R, Castillo A, Cruz Hernández JJ, Grande E. Biomarkers and polymorphisms in pancreatic neuroendocrine tumors treated with sunitinib. Oncotarget 2018; 9:36894-36905. [PMID: 30651923 PMCID: PMC6319342 DOI: 10.18632/oncotarget.26380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022] Open
Abstract
Several circulating biomarkers and single nucleotide polymorphisms (SNPs) have been correlated with efficacy and tolerability to antiangiogenic agents. These associations remain unexplored in well-differentiated, metastatic pancreatic neuroendocrine tumors treated with the multitargeted tyrosine kinase inhibitor sunitinib. We have assessed the effect on tumor response at 6 months, overall survival, progression-free survival and safety of 14 SNPs, and 6 soluble proteins. Forty-three patients were recruited. Two SNPs in the vascular endothelial growth factor receptor 3 (VEGFR-3) gene predicted lower overall survival: rs307826 with hazard ratio (HR) 3.67 (confidence interval [CI] 95%, 1.35-10.00) and rs307821 with HR 3.84 (CI 95%, 1.47-10.0). Interleukin-6 was associated with increased mortality: HR 1.06 (CI 95%, 1.01-1.12), and osteopontin was associated with shorter PFS: HR 1.087 (1.01-1.16), independently of Ki-67. Furthermore, levels of osteopontin remained higher at the end of the study in patients considered non-responders: 38.5 ng/mL vs. responders: 18.7 ng/mL, p-value=0.039. Dynamic upward variations were also observed with respect to IL-8 levels in sunitinib-refractory individuals: 28.5 pg/mL at baseline vs. 38.3 pg/mL at 3 months, p-value=0.024. In conclusion, two VEGFR-3 SNPs as well as various serum biomarkers were associated with diverse clinical outcomes in patients with well-differentiated pancreatic neuroendocrine tumors treated with sunitinib.
Collapse
Affiliation(s)
- Paula Jiménez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Miguel Navarro Martín
- Medical Oncology Department, Hospital Universitario de Salamanca, IBSAL, Salamanca, Spain
| | - Alberto Carmona-Bayonas
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, UMU, IMIB, Murcia, Spain
| | - Alfonso Calvo
- IDISNA and Program in Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), Department of Histology and Pathology, University of Navarra, CIBERONC, ISC-II, Pamplona, Spain
| | - Javier Fernández-Mateos
- Molecular Medicine Unit, IBSAL, Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Miriam Redrado
- IDISNA and Program in Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), Department of Histology and Pathology, University of Navarra, Pamplona, Navarra, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Hospital Universitario Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - Nieves Martínez Lago
- Medical Oncology Department, Hospital Universitario de A Coruña, La Coruña, Spain
| | - Adelaida Lacasta
- Medical Oncology Department, Hospital Universitario Donostia, Guipúzcoa, Spain
| | - Javier Muñarriz
- Medical Oncology Department, Hospital General Universitario de Castellón, Castellón, Spain
| | - Ángel Segura
- Medical Oncology Department, Hospital Universitario La Fe, Valencia, Spain
| | - Josep Fuster
- Medical Oncology Department, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Francisco Barón
- Medical Oncology Department, Hospital Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marta Llanos
- Medical Oncology Department, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Raquel Serrano
- Medical Oncology Department, Hospital Universitario Reina Sofia, Cordoba, Spain
| | - Alfredo Castillo
- Medical Oncology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Enrique Grande
- Medical Oncology Department, MD Anderson Cancer Center, Madrid, Spain
| |
Collapse
|
33
|
Voss MH, Reising A, Cheng Y, Patel P, Marker M, Kuo F, Chan TA, Choueiri TK, Hsieh JJ, Hakimi AA, Motzer RJ. Genomically annotated risk model for advanced renal-cell carcinoma: a retrospective cohort study. Lancet Oncol 2018; 19:1688-1698. [PMID: 30416077 DOI: 10.1016/s1470-2045(18)30648-x] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/15/2018] [Accepted: 08/16/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND The Memorial Sloan Kettering Cancer Center (MSKCC) risk model is an established prognostic tool for metastatic renal-cell carcinoma that integrates clinical and laboratory data, but is agnostic to tumour genomics. Several mutations, including BAP1 and PBRM1, have prognostic value in renal-cell carcinoma. Using two independent clinical trial datasets of patients with metastatic renal-cell carcinoma, we aimed to study whether the addition of the mutation status for several candidate prognostic genes to the MSKCC model could improve the model's prognostic performance. METHODS In this retrospective cohort study, we used available formalin-fixed paraffin-embedded tumour tissue and clinical outcome data from patients with metastatic renal-cell carcinoma assigned to treatment with tyrosine kinase inhibitors in the COMPARZ trial (training cohort; n=357) and RECORD-3 trial (validation cohort; n=258). Eligible patients in both trials were treatment-naive; had histologically confirmed, advanced, or metastatic renal-cell carcinoma; and a Karnofsky performance status score of at least 70. For each cohort, data from patients in all treatment groups (sunitinib and pazopanib in the training cohort, and everolimus and sunitinib in the validation cohort) were pooled for this analysis. In the training cohort, tumour tissue was used to evaluate somatic mutations by next-generation sequencing, and the association between cancer-specific outcomes (overall survival, progression-free survival, and overall response) and the mutation status of six genes of interest (BAP1, PBRM1, TP53, TERT, KDM5C, and SETD2) was tested. Only those genes with prognostic value in this setting were added to the MSKCC risk model to create a genomically annotated version. The validation cohort was used to independently test the prognostic value of the annotated model compared with the original MSKCC risk model. FINDINGS 357 (32%) of 1110 patients assigned to protocol treatment in the COMPARZ study between August, 2008, and September, 2011, were evaluable for mutation status and clinical outcomes in the training cohort. The independent validation cohort included 258 (55%) of 471 evaluable patients, enrolled between October, 2009, and June, 2011, on the RECORD-3 study. In the training cohort, the presence of any mutation in BAP1 or TP53, or both, and absence of any mutation in PBRM1 were prognostic in terms of overall survival (TP53wt/BAP1mut, TP53mut/BAP1wt o TP53mut/BAP1mut vs TP53wt/BAP1wt hazard ratio [HR] 1·57, 95% CI 1·21-2·04; p=0·0008; PBRM1wt vs PBRMmut, HR 1·58, 1·16-2·14; p=0·0035). The mutation status for these three prognostic genes were added to the original MSKCC risk model to create a genomically annotated version. Distribution of participants in the training cohort into the three risk groups of the original MSKCC model changed from 87 (24%) of 357 patients deemed at favourable risk, 217 (61%) at intermediate risk, and 53 (15%) at poor risk, to distribution across four risk groups in the genomically annotated risk model, with 36 (10%) of 357 deemed at favourable risk, 77 (22%) at good risk, 108 (30%) at intermediate risk, and 136 (38%) at poor risk. Addition of genomic information improved model performance for predicting overall survival (C-index: original model, 0·595 [95% CI 0·557-0·634] vs new model, 0·637 [0·595-0·679]) and progression-free survival (0·567 [95% CI 0·529-0·604] vs 0·602 [0·560-0·643]) with adequate discrimination of the proportion of patients who achieved an objective response (Cochran-Armitage one-sided p=0·0014). Analyses in the validation cohort confirmed the superiority of the genomically annotated risk model over the original version. INTERPRETATION The mutation status of BAP1, PBRM1, and TP53 has independent prognostic value in patients with advanced or metastatic renal-cell carcinoma treated with first-line tyrosine kinase inhibitors. Improved stratification of patients across risk groups by use of a genomically annotated model including the mutational status of these three genes warrants further investigation in prospective trials and could be of use as a model to stratify patients with metastatic renal-cell carcinoma in clinical trials. FUNDING Novartis Pharmaceuticals Corporation, MSKCC Support Grant/Core Grant, and the J Randall & Kathleen L MacDonald Research Fund.
Collapse
Affiliation(s)
- Martin H Voss
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Albert Reising
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Yuan Cheng
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Parul Patel
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Mahtab Marker
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Fengshen Kuo
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timothy A Chan
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Toni K Choueiri
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, MA, USA
| | - James J Hsieh
- Washington University in St Louis, St Louis, MO, USA
| | - A Ari Hakimi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
34
|
George DJ, Martini JF, Staehler M, Motzer RJ, Magheli A, Donskov F, Escudier B, Li S, Casey M, Valota O, Laguerre B, Pantuck AJ, Pandha HS, Patel A, Lechuga M, Ravaud A. Phase III Trial of Adjuvant Sunitinib in Patients with High-Risk Renal Cell Carcinoma: Exploratory Pharmacogenomic Analysis. Clin Cancer Res 2018; 25:1165-1173. [PMID: 30401688 DOI: 10.1158/1078-0432.ccr-18-1724] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/28/2018] [Accepted: 11/02/2018] [Indexed: 01/26/2023]
Abstract
PURPOSE In the S-TRAC trial, adjuvant sunitinib prolonged disease-free survival (DFS) versus placebo in patients with loco-regional renal cell carcinoma at high risk of recurrence after nephrectomy. An exploratory analysis evaluated associations between SNPs in several angiogenesis- or hypoxia-related genes and clinical outcomes in S-TRAC. PATIENTS AND METHODS Blood samples were genotyped for 10 SNPs and one insertion/deletion mutation using TaqMan assays. DFS was compared using log-rank tests for each genotype in sunitinib versus placebo groups and between genotypes within each of three (sunitinib, placebo, and combined sunitinib plus placebo) treatment groups. P values were unadjusted. RESULTS In all, 286 patients (sunitinib, n = 142; placebo, n = 144) were genotyped. Longer DFS [HR; 95% confidence interval (CI)] was observed with sunitinib versus placebo for VEGFR1 rs9554320 C/C (HR 0.44; 95% CI, 0.21-0.91; P = 0.023), VEGFR2 rs2071559 T/T (HR 0.46; 95% CI, 0.23-0.90; P = 0.020), and eNOS rs2070744 T/T (HR 0.53; 95% CI, 0.30-0.94; P = 0.028). Shorter DFS was observed for VEGFR1 rs9582036 C/A versus C/C with sunitinib, placebo, and combined therapies (P ≤ 0.05), and A/A versus C/C with sunitinib (P = 0.022). VEGFR1 rs9554320 A/C versus A/A was associated with shorter DFS in the placebo (P = 0.038) and combined (P = 0.006) groups. CONCLUSIONS Correlations between VEGFR1 and VEGFR2 SNPs and longer DFS with sunitinib suggest germline SNPs are predictive of improved outcomes with adjuvant sunitinib in patients with renal cell carcinoma. Independent validation studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Daniel J George
- Department of Medical Oncology, Duke Cancer Center, Durham, North Carolina.
| | | | - Michael Staehler
- Department of Urology, University Hospital of Munich, Munich, Germany
| | - Robert J Motzer
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmed Magheli
- Department of Urology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Frede Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Bernard Escudier
- Department of Urology, Institut Gustave Roussy, Villejuif, France
| | - Sherry Li
- Global Product Development, Pfizer Inc, La Jolla, California
| | - Michelle Casey
- Global Product Development, Pfizer Inc., Collegeville, Pennsylvania
| | - Olga Valota
- Global Product Development, Pfizer S.r.L, Milan, Italy
| | - Brigitte Laguerre
- Department of Medical Oncology, Centre Eugene Marquis, Rennes, France
| | - Allan J Pantuck
- Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles, California
| | - Hardev S Pandha
- Department of Medical Oncology, University of Surrey, Surrey, United Kingdom
| | | | - Maria Lechuga
- Global Product Development, Pfizer S.r.L, Milan, Italy
| | - Alain Ravaud
- Department of Medical Oncology, Bordeaux University Hospital, Bordeaux, France
| |
Collapse
|
35
|
Evaluation of KDR rs34231037 as a predictor of sunitinib efficacy in patients with metastatic renal cell carcinoma. Pharmacogenet Genomics 2018; 27:227-231. [PMID: 28430711 DOI: 10.1097/fpc.0000000000000280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The identification of biomarkers able to predict clinical benefit from vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors is urgently needed. Recently, Maitland and colleagues described an association between KDR-rs34231037 and soluble VEGFR2 levels as well as pazopanib pharmacodynamics. We investigated in a well-characterized series of metastatic clear cell renal cell carcinoma patients whether rs34231037 could influence sunitinib response. Clinical data and DNA were available from an international series of 276 patients. KDR-rs34231037 association with sunitinib response, clinical benefit, and progression-free survival was analyzed using logistic and Cox regression analyses. We found that G-allele carriers were over-represented among patients with clinical benefit during sunitinib treatment compared with those refractory to the treatment (odds ratio: 3.78; 95% confidence interval: 1.02-14.06; P=0.047, multivariable analysis). In conclusion, rs34231037 variant carriers seemed to have better sunitinib response than wild-type patients. Moreover, the association with tumor size reduction suggests that this single nucleotide polymorphism might also identify patients with successful tumor downsizing under anti-VEGFR therapy.
Collapse
|
36
|
Amaya GM, Durandis R, Bourgeois DS, Perkins JA, Abouda AA, Wines KJ, Mohamud M, Starks SA, Daniels RN, Jackson KD. Cytochromes P450 1A2 and 3A4 Catalyze the Metabolic Activation of Sunitinib. Chem Res Toxicol 2018; 31:570-584. [PMID: 29847931 DOI: 10.1021/acs.chemrestox.8b00005] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sunitinib is a multitargeted tyrosine kinase inhibitor associated with idiosyncratic hepatotoxicity. The mechanisms of this toxicity are unknown. We hypothesized that sunitinib undergoes metabolic activation to form chemically reactive, potentially toxic metabolites which may contribute to development of sunitinib-induced hepatotoxicity. The purpose of this study was to define the role of cytochrome P450 (P450) enzymes in sunitinib bioactivation. Metabolic incubations were performed using individual recombinant P450s, human liver microsomal fractions, and P450-selective chemical inhibitors. Glutathione (GSH) and dansylated GSH were used as trapping agents to detect reactive metabolite formation. Sunitinib metabolites were analyzed by liquid chromatography-tandem mass spectrometry. A putative quinoneimine-GSH conjugate (M5) of sunitinib was detected from trapping studies with GSH and dansyl-GSH in human liver microsomal incubations, and M5 was formed in an NADPH-dependent manner. Recombinant P450 1A2 generated the highest levels of defluorinated sunitinib (M3) and M5, with less formation by P450 3A4 and 2D6. P450 3A4 was the major enzyme forming the primary active metabolite N-desethylsunitinib (M1). In human liver microsomal incubations, P450 3A inhibitor ketoconazole reduced formation of M1 by 88%, while P450 1A2 inhibitor furafylline decreased generation of M5 by 62% compared to control levels. P450 2D6 and P450 3A inhibition also decreased M5 by 54 and 52%, respectively, compared to control. In kinetic assays, recombinant P450 1A2 showed greater efficiency for generation of M3 and M5 compared to that of P450 3A4 and 2D6. Moreover, M5 formation was 2.7-fold more efficient in human liver microsomal preparations from an individual donor with high P450 1A2 activity compared to a donor with low P450 1A2 activity. Collectively, these data suggest that P450 1A2 and 3A4 contribute to oxidative defluorination of sunitinib to generate a reactive, potentially toxic quinoneimine. Factors that alter P450 1A2 and 3A activity may affect patient risk for sunitinib toxicity.
Collapse
Affiliation(s)
- Gracia M Amaya
- Department of Pharmaceutical Sciences , Lipscomb University College of Pharmacy and Health Sciences , Nashville , Tennessee 37204-3951 , United States
| | - Rebecca Durandis
- Department of Pharmaceutical Sciences , Lipscomb University College of Pharmacy and Health Sciences , Nashville , Tennessee 37204-3951 , United States
| | - David S Bourgeois
- Department of Pharmaceutical Sciences , Lipscomb University College of Pharmacy and Health Sciences , Nashville , Tennessee 37204-3951 , United States
| | - James A Perkins
- Department of Pharmaceutical Sciences , Lipscomb University College of Pharmacy and Health Sciences , Nashville , Tennessee 37204-3951 , United States
| | - Arsany A Abouda
- Department of Pharmaceutical Sciences , Lipscomb University College of Pharmacy and Health Sciences , Nashville , Tennessee 37204-3951 , United States
| | - Kahari J Wines
- Department of Pharmaceutical Sciences , Lipscomb University College of Pharmacy and Health Sciences , Nashville , Tennessee 37204-3951 , United States
| | - Mohamed Mohamud
- Department of Pharmaceutical Sciences , Lipscomb University College of Pharmacy and Health Sciences , Nashville , Tennessee 37204-3951 , United States
| | - Samuel A Starks
- Department of Pharmaceutical Sciences , Lipscomb University College of Pharmacy and Health Sciences , Nashville , Tennessee 37204-3951 , United States
| | - R Nathan Daniels
- Department of Pharmaceutical Sciences , Lipscomb University College of Pharmacy and Health Sciences , Nashville , Tennessee 37204-3951 , United States.,Department of Pharmacology , Vanderbilt University School of Medicine , Nashville , Tennessee 37232-0146 , United States
| | - Klarissa D Jackson
- Department of Pharmaceutical Sciences , Lipscomb University College of Pharmacy and Health Sciences , Nashville , Tennessee 37204-3951 , United States.,Department of Pharmacology , Vanderbilt University School of Medicine , Nashville , Tennessee 37232-0146 , United States
| |
Collapse
|
37
|
Castellano D, Maroto JP, Espinosa E, Grande E, Bolós MV, Llinares J, Esteban E, González Del Alba A, Climent MA, Arranz JA, Méndez MJ, Fernández Parra E, Antón-Aparicio L, Bayona C, Gallegos I, Gallardo E, Samaniego L, García Donas J. Experience with Sunitinib in metastatic renal cell carcinoma (mRCC) patients: pooled analysis from 3 Spanish observational prospective studies. Expert Opin Drug Saf 2018; 17:573-579. [PMID: 28535693 DOI: 10.1080/14740338.2017.1330410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 05/10/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND A pivotal, randomized, phase III trial demonstrated a statistically significant superiority of sunitinib over interferon-α in metastatic renal cell carcinoma (mRCC) patients. OBJECTIVE To evaluate the effectiveness and safety of sunitinib in patients with advanced or mRCC in routine clinical practice. METHODS Retrospective pooled analysis of clinical data from three observational and prospective studies carried out between 2007 and 2011 in 33 Spanish hospitals. Tumor response, Progression-free survival (PFS) and overall survival (OS), and main sunitinib-related toxicities were registered. RESULTS 224 patients were analyzed. Median PFS 10.6 months (95% CI: 9.02-12.25), median OS 21.9 months (95% CI: 17.2-26.6). Objective response rate (ORR) 43.8% (95% CI: 36.8-50.7). Median time to PR was 3.8 months (95% CI: 3.86-5.99) and to CR 8.2 months (95% CI: 4.75-9.77). The most common ≥ grade-3 AEs were asthenia/fatigue (18.7%), hand-foot syndrome (6.2%), hypertension (5.8%) and neutropenia (4.8%). Hand-foot syndrome, diarrhea and mucositis were confirmed as independent predictors for PFS and/or OS in a multivariate analysis (p < 0.05) Conclusions: Outcomes with sunitinib in daily clinical practice resemble those obtained in clinical trials. Long-term benefit with sunitinib is possible in advanced RCC patients but the appropriate management of toxicities is mandatory to enable patients to remain on treatment.
Collapse
Affiliation(s)
| | - José Pablo Maroto
- b Medical Oncology , Hospital de la Santa Creu i Sant Pau , Barcelona , Spain
| | | | | | | | | | - Emilio Esteban
- f General University Hospital of Asturias , Asturias , Spain
| | | | | | | | - Mª José Méndez
- j Medical Oncology , Hospital Universitario Reina Sofia , Córdoba , Spain
| | | | | | | | | | | | | | - Jesús García Donas
- q 17HM Hospitales, Centro Integral Oncológico HM Clara Campal , Madrid , Spain
| |
Collapse
|
38
|
Berardi R, Torniai M, Partelli S, Rubini C, Pagliaretta S, Savini A, Polenta V, Santoni M, Giampieri R, Onorati S, Barucca F, Murrone A, Bianchi F, Falconi M. Impact of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor (VEGFR) single nucleotide polymorphisms on outcome in gastroenteropancreatic neuroendocrine neoplasms. PLoS One 2018; 13:e0197035. [PMID: 29787601 PMCID: PMC5963762 DOI: 10.1371/journal.pone.0197035] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 04/25/2018] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis represents a key event in cancer development, leading to local invasion e metastatization, and might be considered a basic feature in gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) with a high expression of angiogenic molecules. We aimed to analyze the prognostic and predictive role of angiogenic factors in GEP-NENs through the analysis of single nucleotide polymorphisms (SNPs) of VEGF-A, VEGFR2 and VEGFR3. The genomic DNA of 58 consecutive patients with GEP-NENs treated at our Institution was extracted from peripheral blood. Two SNPs were identified respectively in VEGF-A (rs2010963G>C, rs699947A>C), VEGFR-2 (rs2305948C>T, rs1870377T>A), and VEGFR-3 (rs307821T>C, rs307826C>A) gene. Gene polymorphisms were determined by Real-Time PCR using TaqMan assays. Median age was 57 years (range 24-79 years); 32 patients were male and 77.5% of NENs were localized in the pancreas. The allele frequency of VEGFR-2 rs2305948T and of VEGF-A rs2010963C showed a trend of higher frequency than in general population (12.1% vs. 8.0% and 34.5% vs. 31.2%, respectively). Three out SNPs (VEGF-A rs699947C, VEGF-A rs2010963GC and VEGFR-3 rs307821C) showed a correlation with an increased risk of disease relapse. Moreover median PFS changes according to the presence of 0-1 SNPs (20.7% of cases; 61.9 months), 2 SNPs (25.9%; 49.2 months) and 3 SNPs (53.4%; 27.8 months) (p = 0.034). Results suggest, for the first time, that specific SNPs in VEGF-A and VEGFR-3 correlate with poor prognosis in GEP-NENs. The identification of this new prognostic factor might be helpful in order to optimize the management of these heterogeneous neoplasms.
Collapse
Affiliation(s)
- Rossana Berardi
- Clinica di Oncologia Medica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Mariangela Torniai
- Clinica di Oncologia Medica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Stefano Partelli
- Chirurgia del Pancreas, Ospedale San Raffaele IRCCS, Università Vita e Salute, Milano, Italy
| | - Corrado Rubini
- Section of Pathological Anatomy and Histopathology, Deparment of Neuroscience, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Silvia Pagliaretta
- Clinica di Oncologia Medica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Agnese Savini
- Clinica di Oncologia Medica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Vanessa Polenta
- Dipartimento di Chirurgia Generale, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Matteo Santoni
- Clinica di Oncologia Medica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Riccardo Giampieri
- Clinica di Oncologia Medica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Sofia Onorati
- Clinica di Oncologia Medica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Federica Barucca
- Clinica di Oncologia Medica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Alberto Murrone
- Clinica di Oncologia Medica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Francesca Bianchi
- Clinica di Oncologia Medica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Massimo Falconi
- Section of Pathological Anatomy and Histopathology, Deparment of Neuroscience, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| |
Collapse
|
39
|
Wellmann R, Borden BA, Danahey K, Nanda R, Polite BN, Stadler WM, Ratain MJ, O'Donnell PH. Analyzing the clinical actionability of germline pharmacogenomic findings in oncology. Cancer 2018; 124:3052-3065. [PMID: 29742281 DOI: 10.1002/cncr.31382] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/07/2018] [Accepted: 03/09/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND Germline and tumor pharmacogenomics impact drug responses, but germline markers less commonly guide oncology prescribing. The authors hypothesized that a critical number of clinically actionable germline pharmacogenomic associations exist, representing clinical implementation opportunities. METHODS In total, 125 oncology drugs were analyzed for positive germline pharmacogenomic associations in journals with impact factors ≥5. Studies were assessed for design and genotyping quality, clinically relevant outcomes, statistical rigor, and evidence of drug-gene effects. Associations from studies of high methodologic quality were deemed potentially clinically actionable, and translational summaries were written as point-of-care clinical decision support (CDS) tools and formally evaluated using the Appraisal of Guidelines for Research and Evaluation (AGREE) II instrument. RESULTS The authors identified germline pharmacogenomic results for 56 of 125 oncology drugs (45%) across 173 publications. Actionable associations were detected for 12 drugs, including 6 that had germline pharmacogenomic information within US Food and Drug Administration labels or published guidelines (capecitabine/fluorouracil/dihydropyrimidine dehydrogenase [DPYD], irinotecan/uridine diphosphate glucuronosyltransferase family 1 member A1 [UGT1A1], mercaptopurine/thioguanine/thiopurine S-methyltransferase [TPMT], tamoxifen/cytochrome P450 [CYP] family 2 subfamily D member 6 [CYP2D6]), and 6 others were novel (asparaginase/nuclear factor of activated T-cells 2 [NFATC2]/human leukocyte antigen D-related β1 [HLA-DRB1], cisplatin/acylphosphatase 2 [ACYP2], doxorubicin/adenosine triphosphate-binding cassette subfamily C member 2/Rac family small guanosine triphosphatase 2/neutrophil cytosolic factor 4 [ABCC2/RAC2/NCF4], lapatinib/human leukocyte antigen DQ α1 [HLA-DQA1], sunitinib/cytochrome P450 family 3 subfamily A member 5 [CYP3A5], vincristine/centrosomal protein 72 [CEP72]). By using AGREE II, the developed CDS summaries had high mean ± standard deviation scores (maximum score, 100) for scope and purpose (92.7 ± 5.1) and rigour of development (87.6 ± 7.4) and moderate yet robust scores for clarity of presentation (58.6 ± 25.1) and applicability (55.9 ± 24.6). The overall mean guideline quality score was 5.2 ± 1.0 (maximum score, 7). Germline pharmacogenomic CDS summaries for these 12 drugs were recommended for implementation. CONCLUSIONS Several oncology drugs have actionable germline pharmacogenomic information, justifying their delivery through institutional pharmacogenomic implementations to determine clinical utility. Cancer 2018;124:3052-65. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Rebecca Wellmann
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois
| | - Brittany A Borden
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois
| | - Keith Danahey
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois.,Center for Research Informatics, The University of Chicago, Chicago, Illinois
| | - Rita Nanda
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois.,Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Blase N Polite
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois.,Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Walter M Stadler
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois.,Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Mark J Ratain
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois.,Department of Medicine, The University of Chicago, Chicago, Illinois.,Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois
| | - Peter H O'Donnell
- Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois.,Department of Medicine, The University of Chicago, Chicago, Illinois.,Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois
| |
Collapse
|
40
|
Abstract
Metastatic renal cell carcinoma (mRCC) is an incurable malignancy, characterized by its resistance to traditional chemotherapy, radiation, and hormonal therapy. Treatment perspectives and prognosis of patients with mRCC have been significantly improved by advances in the understanding of its molecular pathogenesis, which have led to the development of targeted therapeutics. Different molecular factors derived from the tumor or the host detected in both tissue or serum could be predictive of therapeutic benefit. Some of them suggest a rational selection of patients to be treated with certain therapies, though none have been validated for routine use. This article provides an overview of both clinical and molecular factors associated with predictive or prognostic value in mRCC and emphasizes that both should be considered in parallel to provide the most appropriate, individualized treatment and achieve the best outcomes in clinical practice.
Collapse
|
41
|
Buonerba C, De Placido P, Bruzzese D, Pagliuca M, Ungaro P, Bosso D, Ribera D, Iaccarino S, Scafuri L, Liotti A, Romeo V, Izzo M, Perri F, Casale B, Grimaldi G, Vitrone F, Brunetti A, Terracciano D, Marinelli A, De Placido S, Di Lorenzo G. Isoquercetin as an Adjunct Therapy in Patients With Kidney Cancer Receiving First-Line Sunitinib (QUASAR): Results of a Phase I Trial. Front Pharmacol 2018; 9:189. [PMID: 29615901 PMCID: PMC5864863 DOI: 10.3389/fphar.2018.00189] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 02/19/2018] [Indexed: 12/14/2022] Open
Abstract
Sunitinib is the most commonly prescribed drug for advanced renal cell carcinoma in the first-line setting and has been associated with multiple adverse events related to its on–and off–target effects, including hand and foot syndrome and fatigue. It was hypothesized that sunitinib-induced fatigue may be related to off target inhibition of the AMPK enzyme, which results in impairment of energy-producing processes at a systemic level. Quercetin is a naturally occurring flavonol with established AMPK-stimulating activity. While clinical use of quercetin is limited by its poor bio-availability, quercetin-3-O-β-d-glucopyranoside, that is isoquercetin, has an improved pharmacokinetic profile. On the grounds of the in vitro stimulatory activity with respect to AMPk, we hypothesized that oral isoquercetin could improve fatigue in kidney cancer patients receiving sunitinib. Given the lack of data on the safety of isoquercetin given concomitantly with sunitinib, we conducted a phase I trial to assess the safety of GMP manufactured isoquercetin given at two dose levels (450 and 900 mg a day). In the 12-patient study cohort included in this study, isoquercetin was administered concomitantly with 50 mg sunitinib for a median 81 days (IQR, 75.5, 86.5). None of the 12 patients required isoquercetin suspension or isoquercetin dose reduction because of adverse events. No abnormalities in ECG, heart or lower limbs doppler ultrasound were detected. A statistically significant improvement was reported for the FACIT fatigue score (6.8 points; 95% CI: 2.8–10.8; p = 0.002) and for the FACIT Adverse Events score (18.9 points; 95% CI: 9.1–28.8; p < 0.001) after isoquercetin consumption vs. baseline. In this phase I trial, isoquercetin was remarkably safe, with a preliminary signal of activity in terms of improvement of sunitinib adverse events.
Collapse
Affiliation(s)
- Carlo Buonerba
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy.,Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Italy
| | - Pietro De Placido
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| | - Dario Bruzzese
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | - Martina Pagliuca
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| | - Paola Ungaro
- Institute of Experimental Endocrinology and Oncology (IEOS-CNR) "G. Salvatore", Naples, Italy
| | - Davide Bosso
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| | - Dario Ribera
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| | - Simona Iaccarino
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| | - Luca Scafuri
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| | - Antonietta Liotti
- Department of Translational Medical Sciences, University "Federico II", Naples, Italy
| | - Valeria Romeo
- Department of Advanced Biomedical Sciences, University Federico II of Naples, Naples, Italy
| | - Michela Izzo
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| | - Francesco Perri
- Medical Oncology Unit, POC SS Annunziata Taranto, Taranto, Italy
| | - Beniamino Casale
- Dipartimento di Pneumologia e Tisiologia, Day Hospital Pneumologia e Pneumoncologico, AORN Vincenzo Monaldi, Naples, Italy
| | - Giuseppe Grimaldi
- U.O. Medicina-Oncoematologia Ospedale Umberto I, Nocera Inferiore, Italy
| | - Francesca Vitrone
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, University Federico II of Naples, Naples, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University "Federico II", Naples, Italy
| | - Alfredo Marinelli
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy.,IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli (IS), Italy
| | - Sabino De Placido
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| | - Giuseppe Di Lorenzo
- Medical Oncology Division, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| |
Collapse
|
42
|
Verbiest A, Lambrechts D, Van Brussel T, Couchy G, Wozniak A, Méjean A, Lerut E, Oudard S, Verkarre V, Job S, de Reynies A, Machiels JP, Patard JJ, Zucman-Rossi J, Beuselinck B. Polymorphisms in the Von Hippel-Lindau Gene Are Associated With Overall Survival in Metastatic Clear-Cell Renal-Cell Carcinoma Patients Treated With VEGFR Tyrosine Kinase Inhibitors. Clin Genitourin Cancer 2018; 16:266-273. [PMID: 29503246 DOI: 10.1016/j.clgc.2018.01.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/17/2018] [Accepted: 01/27/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Clear-cell renal-cell carcinoma (ccRCC) is characterized by loss of a functional Von Hippel-Lindau (VHL) protein. We investigated the potential of 3 single nucleotide polymorphisms (SNPs) in VHL as biomarkers in metastatic ccRCC (m-ccRCC) patients treated with vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs). PATIENTS AND METHODS We genotyped 3 VHL SNPs in 199 m-ccRCC patients: rs1642742 T > C, rs1642743 A > G, and rs1678607 C > A. Primary end points were response rate (RR), progression-free survival (PFS), and overall survival (OS) after start of first-line TKI. RR was compared with Fisher's exact test, and PFS and OS with Kaplan-Meier analysis and multivariable Cox regression. Secondary end points were association with VHL promotor hypermethylation, VHL mutation status, VHL loss of heterozygosity, ≥ 25% sarcomatoid dedifferentiation, and expression of genes implicated in angiogenesis and immunoresponse (Fisher's exact test and unpaired t tests). RESULTS The minor alleles of rs1642742 and rs1642743, known to be in close linkage disequilibrium, were associated with poor outcome, following a recessive pattern. For the rs1642742 CC versus TT/TC genotype, OS was 11 versus 26 months (hazard ratio = 2.3; 95% confidence interval, 1.2-6.6; P = .015). For the rs1642743 GG versus AA/AG genotype, OS was 15 versus 28 months (hazard ratio = 2.6; 95% confidence interval, 1.4-5.0; P = .004). After multivariable analysis, both remained linked with poor OS (P = .018 and P = .009, respectively). There was a trend toward shorter PFS and poorer RR. Both SNPs were associated with ≥ 25% sarcomatoid dedifferentiation (P = .037 and .006, respectively). No significant results were found for rs1678607. CONCLUSION rs1642742 and rs1642743 are candidate biomarkers for poor OS in m-ccRCC patients receiving first-line VEGFR-TKI. They are associated with higher levels of sarcomatoid dedifferentiation.
Collapse
Affiliation(s)
- Annelies Verbiest
- Laboratory of Experimental Oncology, Department of Oncology, University of Leuven, Leuven, Belgium; Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Diether Lambrechts
- Center for Cancer Biology, Flemish Institute for Biotechnology, Leuven, Belgium; Laboratory for Translational Genetics, Department of Human Genetics, University of Leuven, Leuven, Belgium
| | - Thomas Van Brussel
- Center for Cancer Biology, Flemish Institute for Biotechnology, Leuven, Belgium; Laboratory for Translational Genetics, Department of Human Genetics, University of Leuven, Leuven, Belgium
| | - Gabrielle Couchy
- INSERM, UMR-1162, Functional Genomics of Solid Tumors, Institute of Hematology, Paris, France
| | - Agnieszka Wozniak
- Laboratory of Experimental Oncology, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Arnaud Méjean
- Department of Urology, University Hospitalsof Central Paris, Paris, France
| | - Evelyne Lerut
- Department of Pathology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Stéphane Oudard
- Department of Medical Oncology, University Hospitalsof Central Paris, Paris, France
| | - Virginie Verkarre
- Department of Pathology, University Hospitalsof Central Paris, Paris, France
| | - Sylvie Job
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Aurélien de Reynies
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Jean-Pascal Machiels
- Institut Roi Albert II, Service d'Oncologie Médicale, Cliniques universitaires Saint-Luc and Institut de Recherche Clinique et Expérimentale, Université Catholique de Louvain, Brussels, Belgium
| | | | - Jessica Zucman-Rossi
- INSERM, UMR-1162, Functional Genomics of Solid Tumors, Institute of Hematology, Paris, France
| | - Benoit Beuselinck
- Laboratory of Experimental Oncology, Department of Oncology, University of Leuven, Leuven, Belgium; Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium.
| |
Collapse
|
43
|
Bianconi M, Faloppi L, Loretelli C, Zizzi A, Giampieri R, Bittoni A, Andrikou K, Del Prete M, Burattini L, Montironi R, Scartozzi M, Cascinu S. Angiogenesis genotyping in the selection of first-line treatment with either sunitinib or pazopanib for advanced renal cell carcinoma. Oncotarget 2018; 7:37599-37607. [PMID: 27175586 PMCID: PMC5122335 DOI: 10.18632/oncotarget.9229] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/28/2016] [Indexed: 01/21/2023] Open
Abstract
Introduction Recent data from the COMPARZ study seem to suggest a non-inferiority of pazopanib confronted with sunitinib in PFS and OS. We previously reported how VEGF and VEGFR polymorphisms might have a predictive role in patients treated with first-line sunitinib. Aim of our study was to investigate whether tumour angiogenesis genotyping could influence clinical outcome in RCC patients treated with either sunitinib or pazopanib, in order to help clinicians select the appropriate treatment for each patient. Results 19 patients were treated with pazopanib while 78 received sunitinib. VEGF A rs833061 resulted significant in PFS in sunitinib vs pazopanib patients (CC+CT>TT in sunitinib, TT>CC+CT in pazopanib; p<0,0001); VEGF A rs2010963 resulted significant in PFS in sunitinib vs pazopanib patients (GG+CG>CC in sunitinib, CC>GG+CG in pazopanib; p<0,0001); VEGF A rs699947 resulted significant in PFS in sunitinib vs pazopanib patients (AA+AC>CC in sunitinib, CC>AA+AC in pazopanib; p<0,0001). OS showed no statistically significant difference. Conclusions in our analysis patients with opposite polymorphisms of rs833061, rs2010963, rs699947 of VEGF A seems to have a better PFS if treated with either sunitinib or pazopanib. Our data seem to suggest that biology could have a role choosing first line treatment for mRCC patients. Methods a retrospective analysis on 97 histologic samples of mRCC patients was conducted for VEGF-A, VEGF-C and VEGFR-1,2,3 single nucleotide polymorphisms (SNPs).
Collapse
Affiliation(s)
- Maristella Bianconi
- Department of Oncology, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| | - Luca Faloppi
- Department of Oncology, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| | - Cristian Loretelli
- Department of Oncology, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| | - Antonio Zizzi
- Institute of Pathological Anatomy, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| | - Riccardo Giampieri
- Department of Oncology, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| | - Alessandro Bittoni
- Department of Oncology, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| | - Kalliopi Andrikou
- Department of Oncology, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| | - Michela Del Prete
- Department of Oncology, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| | - Luciano Burattini
- Department of Oncology, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| | - Rodolfo Montironi
- Institute of Pathological Anatomy, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| | - Mario Scartozzi
- Department of Medical Oncology, Università degli Studi di Cagliari - Azienda Ospedaliero Universitaria, Cagliari, Italy
| | - Stefano Cascinu
- Department of Oncology, AOU Ospedali Riuniti, Polytechnic University of The Marche Region, Ancona, Italy
| |
Collapse
|
44
|
Yamamoto K, Yano I. Genetic polymorphisms associated with adverse reactions of molecular-targeted therapies in renal cell carcinoma. Med Oncol 2018; 35:16. [PMID: 29302760 DOI: 10.1007/s12032-017-1077-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 12/27/2017] [Indexed: 12/28/2022]
Abstract
The prognosis of patients with metastatic renal cell carcinoma has drastically improved due to the development of molecular-targeted drugs and their use in clinical practice. However, these drugs cause some diverse adverse reactions in patients and sometimes affect clinical outcomes of cancer therapy. Therefore, predictive markers are necessary to avoid severe adverse reactions, to establish novel and effective prevention methods, and to improve treatment outcomes. Some genetic factors involved in these adverse reactions have been reported; however, perspectives on each adverse response have not been integrated yet. In this review, genetic polymorphisms relating to molecular-targeted therapy-induced adverse reactions in patients with renal cell carcinoma are summarized in the points of pharmacokinetic and pharmacodynamic mechanisms. We also discuss about the relationship between systemic drug exposure and adverse drug reactions.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Ikuko Yano
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
45
|
Zhang Y, Mai H, Guo G, Bi G, Hao G, Li Y, Wang X, Cheng L, Wang J, Dong R, Liu Z, Chen L, Qu H. Association analysis of SNPs present in plasma with adverse events and population pharmacokinetics in Chinese sunitinib treated patients with renal cell carcinoma. Oncotarget 2018; 9:14109-14123. [PMID: 29581831 PMCID: PMC5865657 DOI: 10.18632/oncotarget.23881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 11/11/2017] [Indexed: 12/16/2022] Open
Abstract
Background Sunitinib is a tyrosine kinase inhibitor with effective therapeutic outcomes in patients with renal-cell carcinoma. The study were to analyze the association of single-nucleotide polymorphisms present in cell-free DNA and pharmacokinetics with sunitinib treatment-emergent adverse events in Chinese patients with renal-cell carcinoma. Materials and Methods We genotyped 8 keys SNPs in 6 candidate genes. The plasma concentrations of sunitinib and N-desethyl sunitinib were measured using a high performance liquid chromatography-tandam mass spectrometry method. Correlations between the single-nucleotide polymorphisms and adverse events were investigated by univariate and multivariate logistic regression and we quantitatively evaluated the effect of single-nucleotide polymorphisms on the pharmacokinetics of sunitinib by using a population PK model. Results Necessary dose reductions of sunitinib were significantly correlated with SNP rs1933437 in FLT3. A higher severity of AEs were collected with SNP rs2032582 in ABCB1 and rs1800812 in PDGFRα. Thrombocytopenia was collected with rs1800812 in PDGFRα. Our study provides a population PK model of sunitinib with the ABCB1 genotype as a predictive covariate for apparent oral clearance. Conclusions Our study preliminarily confirmed the hypothesis that the pharmacokinetics of sunitinib is affected by the SNPs of enzyme in Chinese renal-cell carcinoma patients, and this affects the different distribution and severity of adverse events of sunitinib.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Haixing Mai
- Department of Urology Department, Academy of Military Medical Sciences Affiliated Hospital, Beijing 100071, China
| | - Gang Guo
- Department of Urology Department, The General Hospital of the People's Liberation Army, Beijing 100853, China
| | - Guofang Bi
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Guangtao Hao
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Yuanyuan Li
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Xiaofang Wang
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Longmei Cheng
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Jing Wang
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Ruihua Dong
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Zeyuan Liu
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Lijun Chen
- Department of Urology Department, Academy of Military Medical Sciences Affiliated Hospital, Beijing 100071, China
| | - Hengyan Qu
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| |
Collapse
|
46
|
Tornio A, Backman JT. Cytochrome P450 in Pharmacogenetics: An Update. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2018; 83:3-32. [PMID: 29801580 DOI: 10.1016/bs.apha.2018.04.007] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Interindividual variability in drug disposition is a major cause of lack of efficacy and adverse effects of drug therapies. The majority of hepatically cleared drugs are metabolized by cytochrome P450 (CYP) enzymes, mainly in families CYP1, CYP2, and CYP3. Genes encoding these enzymes are highly variable with allele distribution showing considerable differences between populations. Genetic variability of especially CYP2C9, CYP2C19, CYP2D6, and CYP3A5 is known to have clear clinical impact on drugs that are metabolized by these enzymes. CYP1A2, CYP2A6, CYP2B6, CYP2C8, and CYP3A4 all show variability that affects pharmacokinetics of drugs as well, but so far the evidence regarding their clinical implications is not as conclusive. In this review, we provide an up-to-date summary of the pharmacogenetics of the major human drug-metabolizing CYP enzymes, focusing on clinically significant examples.
Collapse
Affiliation(s)
- Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
47
|
Garrigós C, Espinosa M, Salinas A, Osman I, Medina R, Taron M, Molina-Pinelo S, Duran I. Single nucleotide polymorphisms as prognostic and predictive biomarkers in renal cell carcinoma. Oncotarget 2017; 8:106551-106564. [PMID: 29290970 PMCID: PMC5739755 DOI: 10.18632/oncotarget.22533] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 10/25/2017] [Indexed: 11/25/2022] Open
Abstract
Despite major advances in the knowledge of the molecular basis of renal cell carcinoma, prognosis is still defined using clinical and pathological parameters. Moreover, no valid predictive biomarkers exist to help us selecting the best treatment for each patient. With these premises, we aimed to analyse the expression and to determine the prognostic and predictive value of 64 key single nucleotide polymorphisms in 18 genes related with angiogenesis or metabolism of antiangiogenics in two cohorts of patients with localized and advanced renal cell cancer treated at our institution. The presence of the selected single nucleotide polymorphisms was correlated with clinical features, disease free survival, overall survival and response rate. In patients with localized renal cell cancer, 5 of these polymorphisms in 3 genes involved in angiogenesis predicted for worse disease free survival (VEGFR2: rs10013228; PDGFRA: rs2228230) or shorter overall survival (VEGFR2: rs10013228; VEGFR3: rs6877011, rs307826) (p < 0.05). Rs2071559 in VEGFR2 showed a protective effect (p = 0.01). In the advanced setting, 5 SNPs determined inferior overall survival (IL8: rs2227543, PRKAR1B: rs9800958, PDGFRB: rs2302273; p = 0.05) or worse response rate (VEGFA: rs699947, rs3025010 p ≤ 0.01)). Additionally 1 single nucleotide polymorphism in VEGFB predicted for better response rate rs594942 (p = 0.03). Genetic analysis of renal cell carcinoma patients might provide valuable prognostic/predictive information. A set of SNPs in genes critical to angiogenesis and metabolism of antiangiogenics drugs seem to determine post-surgical outcomes and treatment response in our series.
Collapse
Affiliation(s)
- Carmen Garrigós
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Marta Espinosa
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Ana Salinas
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Ignacio Osman
- Unidad de Urología Oncológica, UGC Urología-Nefrología H.U.Virgen del Rocío, Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Rafael Medina
- Unidad de Urología Oncológica, UGC Urología-Nefrología H.U.Virgen del Rocío, Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Miguel Taron
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Sonia Molina-Pinelo
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Sevilla, Spain.,Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain
| | - Ignacio Duran
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
48
|
Duran I, Lambea J, Maroto P, González-Larriba JL, Flores L, Granados-Principal S, Graupera M, Sáez B, Vivancos A, Casanovas O. Resistance to Targeted Therapies in Renal Cancer: The Importance of Changing the Mechanism of Action. Target Oncol 2017; 12:19-35. [PMID: 27844272 DOI: 10.1007/s11523-016-0463-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Renal cell carcinoma (RCC) is a complex disease characterized by mutations in several genes. Loss of function of the von Hippel-Lindau (VHL) tumour suppressor gene is a very common finding in RCC and leads to up-regulation of hypoxia-inducible factor (HIF)-responsive genes accountable for angiogenesis and cell growth, such as platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF). Binding of these proteins to their cognate tyrosine kinase receptors on endothelial cells promotes angiogenesis. Promotion of angiogenesis is in part due to the activation of the phosphatidylinositol-3-kinase (PI3K)/AKT/mechanistic target of rapamycin (mTOR) pathway. Inhibition of this pathway decreases protein translation and inhibits both angiogenesis and tumour cell proliferation. Although tyrosine kinase inhibitors (TKIs) stand as the main first-line treatment option for advanced RCC, eventually all patients will become resistant to TKIs. Resistance can be overcome by using second-line treatments with different mechanisms of action, such as inhibitors of mTOR, c-MET, programmed death 1 (PD-1) receptor, or the combination of an mTOR inhibitor (mTORi) with a TKI. In this article, we briefly review current evidence regarding mechanisms of resistance in RCC and treatment strategies to overcome resistance with a special focus on the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- I Duran
- Sección de Oncología Médica, Hospital Universitario Virgen del Rocío, Sevilla, Spain.,Laboratorio de Terapias Avanzadas y Biomarcadores en Oncología, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - J Lambea
- Servicio de Oncología Médica, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - P Maroto
- Servicio de Oncología Médica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | - S Granados-Principal
- Servicio de Oncología Médica, Complejo Hospitalario de Jaén, Jaén, Spain.,GENYO, Centre for Genomics and Oncological Research (Pfizer/University of Granada/Andalusian Regional Government), PTS Granada, Granada, Spain
| | - M Graupera
- Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, Barcelona, Spain
| | - B Sáez
- Departmento de Bioquímica, Biología Molecular y Celular, Instituto Universitario de Investigación en Nanociencia de Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - A Vivancos
- Departamento de Bioquímica y Biología Molecular, Universidad Pompeu Fabra, Barcelona, Spain
| | - O Casanovas
- ProCURE Research Program, Institut Català d'Oncologia-IDIBELL, L'Hospitalet de Llobregat, Avinguda Gran Via, 199-203, 08907, Barcelona, Spain.
| |
Collapse
|
49
|
Motzer RJ, Figlin RA, Martini JF, Hariharan S, Agarwal N, Li CX, Williams JA, Hutson TE. Germline Genetic Biomarkers of Sunitinib Efficacy in Advanced Renal Cell Carcinoma: Results From the RENAL EFFECT Trial. Clin Genitourin Cancer 2017; 15:526-533. [DOI: 10.1016/j.clgc.2017.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/10/2017] [Accepted: 02/19/2017] [Indexed: 10/20/2022]
|
50
|
Li M, Kroetz DL. Bevacizumab-induced hypertension: Clinical presentation and molecular understanding. Pharmacol Ther 2017; 182:152-160. [PMID: 28882537 DOI: 10.1016/j.pharmthera.2017.08.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bevacizumab is a vascular endothelial growth factor-A-specific angiogenesis inhibitor indicated as an adjunct to chemotherapy for the treatment of several types of cancer. Hypertension is commonly observed during bevacizumab treatment, and high-grade toxicity can limit therapy and lead to other cardiovascular complications. The factors that contribute to interindividual variability in blood pressure response to bevacizumab treatment are not well understood. In this review, we outline research efforts to understand the mechanisms and pathophysiology of hypertension resulting from bevacizumab treatment. Moreover, we highlight current knowledge of the pharmacogenetics of bevacizumab-induced hypertension, which may be used to develop strategies to prevent or minimize this toxicity.
Collapse
Affiliation(s)
- Megan Li
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Deanna L Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States.
| |
Collapse
|