1
|
Abad A, Martínez-Balibrea E, Viéitez J, Alonso-Orduña V, García Alfonso P, Manzano J, Massutí B, Benavides M, Carrato A, Zanui M, Gallego J, Grávalos C, Conde V, Provencio M, Valladares-Ayerbes M, Salazar R, Sastre J, Montagut C, Rivera F, Aranda E. Genotype-based selection of treatment of patients with advanced colorectal cancer (SETICC): a pharmacogenetic-based randomized phase II trial. Ann Oncol 2018; 29:439-444. [DOI: 10.1093/annonc/mdx737] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
2
|
Kumuthini J, Mbiyavanga M, Chimusa ER, Pathak J, Somervuo P, Van Schaik RH, Dolzan V, Mizzi C, Kalideen K, Ramesar RS, Macek M, Patrinos GP, Squassina A. Minimum information required for a DMET experiment reporting. Pharmacogenomics 2016; 17:1533-45. [PMID: 27548815 DOI: 10.2217/pgs-2016-0015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIM To provide pharmacogenomics reporting guidelines, the information and tools required for reporting to public omic databases. MATERIAL & METHODS For effective DMET data interpretation, sharing, interoperability, reproducibility and reporting, we propose the Minimum Information required for a DMET Experiment (MIDE) reporting. RESULTS MIDE provides reporting guidelines and describes the information required for reporting, data storage and data sharing in the form of XML. CONCLUSION The MIDE guidelines will benefit the scientific community with pharmacogenomics experiments, including reporting pharmacogenomics data from other technology platforms, with the tools that will ease and automate the generation of such reports using the standardized MIDE XML schema, facilitating the sharing, dissemination, reanalysis of datasets through accessible and transparent pharmacogenomics data reporting.
Collapse
Affiliation(s)
- Judit Kumuthini
- Centre for Proteomic & Genomic Research, Cape Town, South Africa
| | | | - Emile R Chimusa
- Centre for Proteomic & Genomic Research, Cape Town, South Africa.,Computational Biology Group, Institute for Infectious Diseases & Molecular Medicine, University of Cape Town, South Africa
| | - Jyotishman Pathak
- Division of Biomedical Statistics & Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Panu Somervuo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ron Hn Van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Center Rotterdam, Room Na-415, Wytemaweg 80, 3015CN Rotterdam, The Netherlands
| | - Vita Dolzan
- Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Clint Mizzi
- Department of Bioinformatics, Faculty of Medicine & Health Sciences, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Physiology & Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta
| | - Kusha Kalideen
- UCT/SA MRC Human Genetics Research Unit, Division of Human Genetics, Institute for Infectious Diseases & Molecular Medicine, Division of Human Genetics, University of Cape Town, South Africa
| | - Raj S Ramesar
- UCT/SA MRC Human Genetics Research Unit, Division of Human Genetics, Institute for Infectious Diseases & Molecular Medicine, Division of Human Genetics, University of Cape Town, South Africa
| | - Milan Macek
- Department of Biology & Medical Genetics, Charles University Prague & 2nd Faculty of Medicine, Prague, Czechia
| | - George P Patrinos
- Department of Bioinformatics, Faculty of Medicine & Health Sciences, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
| | - Alessio Squassina
- Laboratory of Pharmacogenomics, Section of Neuroscience & Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, sp 8 Sestu-Monserrato, Km 0.700, 09042 Cagliari, Italy
| |
Collapse
|
3
|
Song Q, Zhou X, Yu J, Dong N, Wang X, Yang H, Ren J, Kim Lyerly H. The prognostic values of CYP2B6 genetic polymorphisms and metastatic sites for advanced breast cancer patients treated with docetaxel and thiotepa. Sci Rep 2015; 5:16775. [PMID: 26602960 PMCID: PMC4658481 DOI: 10.1038/srep16775] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/19/2015] [Indexed: 11/09/2022] Open
Abstract
This study investigated interactive effects of CYP2B6 genotypes and liver metastasis on the prognosis of metastatic breast cancer patients who received combined chemotherapy of docetaxel and thiotepa. Totally 153 patients were retrospectively genotyped rs8192719 (c.1294 + 53C > T) and rs2279343 (c.785A > G). Kaplan-Meier method and Cox Proportional Hazard Regression model were used to estimate the survival. Patients with liver metastasis had worsen prognosis, conferring a 2.26-fold high risk of progression and 1.93-fold high risk of death (p < 0.05). Both CT/TT genotype of rs8192719 (c.1294 + 3C > T) and AG genotype of rs2279343 (c.785A > G) prolonged survival (p < 0.05). Furthermore, among liver metastatic patients, AG genotype of rs2279343 (c.785A > G) was associated with a 47% reduced risk of death and a 6-month-longer overall survival (p < 0.05). Among non-liver metastatic patients, hazard ratios of CT/TT genotype of rs8192719 (c.1294 + 53C > T) were 0.45 for progression and 0.40 for death; and the corresponding survival was improved by 6 months and 16 months, respectively (p < 0.05). Genotypes of CYP2B6 had an interaction with clinical efficacy of docetaxel and thiotepa on metastatic breast cancer patients; and metastatic sites also affected clinical responses. Further therapies should take into account of chemotherapy regimen, genotypes of metabolizing enzymes and metastatic sites for the particular subpopulation.
Collapse
Affiliation(s)
- Qingkun Song
- Beijing Key Laboratory of Therapeutic Cancer Vaccines, Beijing Shijitan Hospital, Capital Medical University Cancer Center, 10 Tieyi Road, Beijing 100038, China
| | - Xinna Zhou
- Beijing Key Laboratory of Therapeutic Cancer Vaccines, Beijing Shijitan Hospital, Capital Medical University Cancer Center, 10 Tieyi Road, Beijing 100038, China
| | - Jing Yu
- Department of Medical Oncology, Peking University Cancer Hospital & Institute. 52 Fucheng Rd, Beijing 100142, China
| | - Ningning Dong
- Department of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, Beijing 100050, China
| | - Xiaoli Wang
- Beijing Key Laboratory of Therapeutic Cancer Vaccines, Beijing Shijitan Hospital, Capital Medical University Cancer Center, 10 Tieyi Road, Beijing 100038, China
| | - Huabing Yang
- Beijing Key Laboratory of Therapeutic Cancer Vaccines, Beijing Shijitan Hospital, Capital Medical University Cancer Center, 10 Tieyi Road, Beijing 100038, China
| | - Jun Ren
- Beijing Key Laboratory of Therapeutic Cancer Vaccines, Beijing Shijitan Hospital, Capital Medical University Cancer Center, 10 Tieyi Road, Beijing 100038, China
- Department of Surgery, Duke University Medical Center, 203 Research Drive, Suite 433, Box 2606, Durham, NC 27710, United States
| | - H Kim Lyerly
- Department of Surgery, Duke University Medical Center, 203 Research Drive, Suite 433, Box 2606, Durham, NC 27710, United States
| |
Collapse
|
4
|
Milowsky MI, O'Donnell PH, Flaig TW, Theodorescu D. Molecular determinants of chemotherapy response. Bladder Cancer 2015. [DOI: 10.1002/9781118674826.ch24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
5
|
Are pharmacogenomic biomarkers an effective tool to predict taxane toxicity and outcome in breast cancer patients? Literature review. Cancer Chemother Pharmacol 2015. [PMID: 26198313 DOI: 10.1007/s00280-015-2818-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Breast cancer is a heterogeneous disease, characterized by various molecular phenotypes that correlate with different prognosis and response to treatments. Taxanes are some of the most active chemotherapeutic agents for breast cancer; however, their utilization is limited, due to hematologic and cumulative neurotoxicity on treated patients. To understand why only some patients experience severe adverse effects and why patients respond and develop resistance with different rates to taxane therapy, the metabolic pathways of these drugs should be completely unraveled. The variant forms of several genes, related to taxane pharmacokinetics, can be indicative markers of clinical parameters, such as toxicity or outcome. METHODS The search of the data has been conducted through PubMed database, presenting clinical data, clinical trials and basic research restricted to English language until June 2015. RESULTS We studied the literature in order to find any possible association between the major pharmacogenomic variants and specific taxane-related toxicity and patient outcome. We found that the data of these studies are sometimes discordant, due to both the small number of enrolled patients and the heterogeneity of the examined population. CONCLUSIONS Among all analyzed genes, only CYP1B1 and ABCB1 resulted the strongest candidates to become biomarkers of clinical response to taxane therapy in breast cancer, although their utilization still remains an experimental procedure. In the future, greater studies on genetic polymorphisms should be performed in order to identify differentiating signatures for patients with higher toxicity and with resistant or responsive outcome, before the administration of taxanes.
Collapse
|
6
|
Clarke JD, Hardwick RN, Lake AD, Canet MJ, Cherrington NJ. Experimental nonalcoholic steatohepatitis increases exposure to simvastatin hydroxy acid by decreasing hepatic organic anion transporting polypeptide expression. J Pharmacol Exp Ther 2014; 348:452-8. [PMID: 24403518 DOI: 10.1124/jpet.113.211284] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Simvastatin (SIM)-induced myopathy is a dose-dependent adverse drug reaction (ADR) that has been reported to occur in 18.2% of patients receiving a 40- to 80-mg dose. The pharmacokinetics of SIM hydroxy acid (SIMA), the bioactive form of SIM, and the occurrence of SIM-induced myopathy are linked to the function of the organic anion transporting polypeptide (Oatp) hepatic uptake transporters. Genetic polymorphisms in SLCO1B1, the gene for human hepatic OATP1B1, cause decreased elimination of SIMA and increased risk of developing myopathy. Nonalcoholic steatohepatitis (NASH) is the most severe form of nonalcoholic fatty liver disease, and is known to alter drug transporter expression and drug disposition. The purpose of this study was to assess the metabolism and disposition of SIM in a diet-induced rodent model of NASH. Rats were fed a methionine- and choline-deficient diet for 8 weeks to induce NASH and SIM was administered intravenously. Diet-induced NASH caused increased plasma retention and decreased biliary excretion of SIMA due to decreased protein expression of multiple hepatic Oatps. SIM exhibited increased volume of distribution in NASH as evidenced by increased muscle, decreased plasma, and no change in biliary concentrations. Although Cyp3a and Cyp2c11 proteins were decreased in NASH, no alterations in SIM metabolism were observed. These data, in conjunction with our previous data showing that human NASH causes a coordinated downregulation of hepatic uptake transporters, suggest that NASH-mediated transporter regulation may play a role in altered SIMA disposition and the occurrence of myopathy.
Collapse
Affiliation(s)
- John D Clarke
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona
| | | | | | | | | |
Collapse
|
7
|
Jiang WQ, Fu FF, Li YX, Wang WB, Wang HH, Jiang HP, Teng LS. Molecular biomarkers of colorectal cancer: prognostic and predictive tools for clinical practice. J Zhejiang Univ Sci B 2013; 13:663-75. [PMID: 22949358 DOI: 10.1631/jzus.b1100340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Colorectal cancer remains one of the most common types of cancer and leading causes of cancer death worldwide. Although we have made steady progress in chemotherapy and targeted therapy, evidence suggests that the majority of patients undergoing drug therapy experience severe, debilitating, and even lethal adverse drug events which considerably outweigh the benefits. The identification of suitable biomarkers will allow clinicians to deliver the most appropriate drugs to specific patients and spare them ineffective and expensive treatments. Prognostic and predictive biomarkers have been the subjects of many published papers, but few have been widely incorporated into clinical practice. Here, we want to review recent biomarker data related to colorectal cancer, which may have been ready for clinical use.
Collapse
Affiliation(s)
- Wei-qin Jiang
- Cancer Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | | | | | | | | | | | | |
Collapse
|
8
|
Crona D, Innocenti F. Can knowledge of germline markers of toxicity optimize dosing and efficacy of cancer therapy? Biomark Med 2012; 6:349-62. [PMID: 22731909 DOI: 10.2217/bmm.12.19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The systemic treatment of cancer with traditional cytotoxic chemotherapeutic agents and more targeted agents is often complicated by the onset of adverse drug reactions. Pharmacogenetic prediction of adverse drug reactions might have consequences for dosing and efficacy. This review discusses relevant examples where the germline variant-toxicity relationship has been validated as an initial step in developing clinically useful pharmacogenetic markers and provides examples where germline variants have influenced dosing strategies and/or survival or other outcomes of efficacy. This review will also provide insight into the reasons why more pharmacogenetic markers have not been routinely integrated into clinical practice.
Collapse
Affiliation(s)
- Daniel Crona
- Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, USA
| | | |
Collapse
|
9
|
Patterns of cancer genetic testing: a randomized survey of Oregon clinicians. J Cancer Epidemiol 2012; 2012:294730. [PMID: 23150730 PMCID: PMC3485974 DOI: 10.1155/2012/294730] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 11/17/2022] Open
Abstract
Introduction. Appropriate use of genetic tests for population-based cancer screening, diagnosis of inherited cancers, and guidance of cancer treatment can improve health outcomes. We investigated clinicians' use and knowledge of eight breast, ovarian, and colorectal cancer genetic tests. Methods. We conducted a randomized survey of 2,191 Oregon providers, asking about their experience with fecal DNA, OncoVue, BRCA, MMR, CYP2D6, tumor gene expression profiling, UGT1A1, and KRAS. Results. Clinicians reported low confidence in their knowledge of medical genetics; most confident were OB-GYNs and specialists. Clinicians were more likely to have ordered/recommended BRCA and MMR than the other tests, and OB-GYNs were twice as likely to have ordered/recommended BRCA testing than primary care providers. Less than 10% of providers ordered/recommended OncoVue, fecal DNA, CYP2D6, or UGT1A1; less than 30% ordered/recommended tumor gene expression profiles or KRAS. The most common reason for not ordering/recommending these tests was lack of familiarity. Conclusions. Use of appropriate, evidence-based testing can help reduce incidence and mortality of certain cancers, but these tests need to be better integrated into clinical practice. Continued evaluation of emerging technologies, dissemination of findings, and an increase in provider confidence and knowledge are necessary to achieve this end.
Collapse
|
10
|
Dong N, Yu J, Wang C, Zheng X, Wang Z, Di L, Song G, Zhu B, Che L, Jia J, Jiang H, Zhou X, Wang X, Ren J. Pharmacogenetic assessment of clinical outcome in patients with metastatic breast cancer treated with docetaxel plus capecitabine. J Cancer Res Clin Oncol 2012; 138:1197-203. [DOI: 10.1007/s00432-012-1183-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 02/21/2012] [Indexed: 10/28/2022]
|
11
|
O'Donnell PH, Stadler WM. Pharmacogenomics in early-phase oncology clinical trials: is there a sweet spot in phase II? Clin Cancer Res 2012; 18:2809-16. [PMID: 22427349 DOI: 10.1158/1078-0432.ccr-11-2445] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Many clinical trials of oncology drugs now include at least a consideration of pharmacogenomics, the study of germline or acquired genetic factors governing a drug's response and toxicity. Besides the potential benefit to patients from the consideration of personalized pharmacogenomic information when making treatment decisions, the incentive is clear for oncology drug developers to incorporate pharmacogenomic factors in the drug development process, because pharmacogenomic biomarkers may allow predictive characterization of subpopulations within a disease that may particularly respond or may allow preidentification of patients at highest risk for adverse events. There is, however, a lack of agreement in actual practice about at what point in the oncology clinical drug development process pharmacogenomic studies should be incorporated. In this article, we examine the recent growth of pharmacogenomics in oncology clinical trials, especially in early-phase studies, and examine several critical questions facing the incorporation of pharmacogenomics in early oncologic drug development. We show that phase II clinical trials, in particular, have a favorable track record for showing positive pharmacogenomic signals, worthy of additional follow-up and validation, and that the phase II setting holds significant promise for potentially accelerating and informing future phase III trials. We conclude that phase II trials offer an ideal "sweet spot" for routine incorporation of pharmacogenomic questions in oncology drug development.
Collapse
Affiliation(s)
- Peter H O'Donnell
- Section of Hematology/Oncology, Department of Medicine, and Comprehensive Cancer Center Pharmacogenomics and Experimental Therapeutics Program, The University of Chicago, Chicago, Illinois, USA.
| | | |
Collapse
|
12
|
Germline pharmacogenomics in oncology: decoding the patient for targeting therapy. Mol Oncol 2012; 6:251-9. [PMID: 22321460 DOI: 10.1016/j.molonc.2012.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 12/14/2011] [Accepted: 01/13/2012] [Indexed: 11/24/2022] Open
Abstract
Pharmacogenomics is the study of genetic factors determining drug response or toxicity. The use of pharmacogenomics is especially desirable in oncology because the therapeutic index of oncology drugs is often narrow, the need for favorable drug response is often acute, and the consequences of drug toxicity can be life-threatening. In this review, we examine the state of pharmacogenomics in oncology, focusing only on germline pharmacogenomic variants. We consider several critical points when assessing the quality of pharmacogenomic findings and their relevance to clinical use, and discuss potential confounding factors limiting interpretation and implementation. Several of the most extensively studied drug-gene pairs (irinotecan and UGT1A1; tamoxifen and CYP2D6; 5-fluorouracil and DPYD) are inspected in depth as illustrations of both the state of advancement-and the current limitations of-present knowledge. We argue that there will likely soon be a critical mass of important germline pharmacogenomic biomarkers in oncology which deserve clinical implementation to provide optimal, personalized oncologic care. We conclude with a vision of how routine clinical testing of such germline markers could one day change the paradigm for cancer care.
Collapse
|
13
|
Nicholson JK, Everett JR, Lindon JC. Longitudinal pharmacometabonomics for predicting patient responses to therapy: drug metabolism, toxicity and efficacy. Expert Opin Drug Metab Toxicol 2012; 8:135-9. [DOI: 10.1517/17425255.2012.646987] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|