1
|
Sarhangi N, Rouhollah F, Niknam N, Sharifi F, Nikfar S, Larijani B, Patrinos GP, Hasanzad M. Pharmacogenetic DPYD allele variant frequencies: A comprehensive analysis across an ancestrally diverse Iranian population. Daru 2024; 32:715-727. [PMID: 39424756 PMCID: PMC11555172 DOI: 10.1007/s40199-024-00538-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/24/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Cancer treatment has improved over the past decades, but many cancer patients still experience adverse drug reactions (ADRs). Pharmacogenomics (PGx), known as personalized treatment, is a pillar of precision medicine that aims to optimize the efficacy and safety of medications by studying the germline variations. Germline variations in the DPYD lead to significant ADRs. The present cross-sectional study aims to evaluate the allele frequency of the DPYD gene variations in the Iranian population to provide insights into personalized treatment decisions in the Iranian population. METHODS The allele frequency of 51 pharmacogenetic variations in the clinically relevant DPYD was assessed in a representative sample set of 1142 unrelated Iranian individuals and subpopulations of different ethnic groups who were genotyped using the Infinium Global Screening Array-24 BeadChip. RESULTS The genotyping assay revealed eight pharmacogenetic variants including DPYD rs1801265 (c.85T > C; DPYD*9A), rs2297595 (c.496A > G), rs1801158 (c.1601G > A; DPYD*4), rs1801159 (c.1627A > G; DPYD*5), rs1801160 (c.2194G > A; DPYD*6), rs17376848 (c.1896T > C), rs56038477 (c.1236G > A; HapB3), and rs75017182 (c.1129-5923C > G; HapB3) with minor allele frequency (MAF) ≥ 1%. CONCLUSION The results of the study reveal significant genetic variations among Iranian population that could significantly influence clinical decision-making. These variants, with their potential to explain the substantial variability in drug response phenotypes among different populations, shed light on a crucial aspect of pharmacogenomics. These findings not only provide valuable insights but also inspire the design and implementation of future pharmacogenomic clinical trials, motivating further research in this crucial area.
Collapse
Affiliation(s)
- Negar Sarhangi
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, 1916893813, Iran
| | - Fatemeh Rouhollah
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, 1916893813, Iran
| | - Negar Niknam
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
- LifeandMe, Inc., Tehran, 1497719825, Iran
| | - Farshad Sharifi
- Elderly Health Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713119, Iran
| | - Shekoufeh Nikfar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713119, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713119, Iran
| | - George P Patrinos
- School of Health Sciences, Department of Pharmacy, University of Patras, Patras, Greece
- College of Medicine and Health Sciences, Department of Genetics and Genomics, United Arab Emirates University, Al-Ain, Abu Dhabi, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain, Abu Dhabi, UAE
| | - Mandana Hasanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, 1411713119, Iran.
| |
Collapse
|
2
|
Reizine N, O’Donnell PH. Modern developments in germline pharmacogenomics for oncology prescribing. CA Cancer J Clin 2022; 72:315-332. [PMID: 35302652 PMCID: PMC9262778 DOI: 10.3322/caac.21722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/15/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
The integration of genomic data into personalized treatment planning has revolutionized oncology care. Despite this, patients with cancer remain vulnerable to high rates of adverse drug events and medication inefficacy, affecting prognosis and quality of life. Pharmacogenomics is a field seeking to identify germline genetic variants that contribute to an individual's unique drug response. Although there is widespread integration of genomic information in oncology, somatic platforms, rather than germline biomarkers, have dominated the attention of cancer providers. Patients with cancer potentially stand to benefit from improved integration of both somatic and germline genomic information, especially because the latter may complement treatment planning by informing toxicity risk for drugs with treatment-limiting tolerabilities and narrow therapeutic indices. Although certain germline pharmacogenes, such as TPMT, UGT1A1, and DPYD, have been recognized for decades, recent attention has illuminated modern potential dosing implications for a whole new set of anticancer agents, including targeted therapies and antibody-drug conjugates, as well as the discovery of additional genetic variants and newly relevant pharmacogenes. Some of this information has risen to the level of directing clinical action, with US Food and Drug Administration label guidance and recommendations by international societies and governing bodies. This review is focused on key new pharmacogenomic evidence and oncology-specific dosing recommendations. Personalized oncology care through integrated pharmacogenomics represents a unique multidisciplinary collaboration between oncologists, laboratory science, bioinformatics, pharmacists, clinical pharmacologists, and genetic counselors, among others. The authors posit that expanded consideration of germline genetic information can further transform the safe and effective practice of oncology in 2022 and beyond.
Collapse
Affiliation(s)
- Natalie Reizine
- Division of Hematology and Oncology, Department of Medicine, The University of Illinois at Chicago
| | - Peter H. O’Donnell
- Section of Hematology/Oncology, Department of Medicine, Center for Personalized Therapeutics, and Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago
- Correspondence to: Dr. Peter H. O’Donnell, Section of Hematology/Oncology, Department of Medicine, The University of Chicago, 5841 S. Maryland Avenue, MC2115, Chicago, IL 60637, USA. ()
| |
Collapse
|
3
|
Reizine NM, Danahey K, Truong TM, George D, House LK, Karrison TG, van Wijk XMR, Yeo KTJ, Ratain MJ, O'Donnell PH. Clinically actionable genotypes for anticancer prescribing among >1500 patients with pharmacogenomic testing. Cancer 2022; 128:1649-1657. [PMID: 35090043 PMCID: PMC9153953 DOI: 10.1002/cncr.34104] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND In recent years, there has been increasing evidence supporting the role of germline pharmacogenomic factors predicting toxicity for anticancer therapies. Although somatic genomic data are used frequently in oncology care planning, germline pharmacogenomic testing is not. This study hypothesizes that comprehensive germline pharmacogenomic profiling could have high relevance for cancer care. METHODS Between January 2011 and August 2020, patients at the University of Chicago Medical Center were genotyped across custom germline pharmacogenomic panels for reasons unrelated to cancer care. Actionable anticancer pharmacogenomic gene/drug interactions identified by the FDA were defined including: CYP2C9 (erdafitinib), CYP2D6 (gefitinib), DPYD (5-fluorouracil and capecitabine), TPMT (thioguanine and mercaptopurine), and UGT1A1 (belinostat, irinotecan, nilotinib, pazopanib, and sacituzumab-govitecan hziy). The primary objective was to determine the frequency of individuals with actionable or high-risk genotypes across these 5 key pharmacogenes, thus potentially impacting prescribing for at least 1 of these 11 commonly prescribed anticancer therapies. RESULTS Data from a total of 1586 genotyped individuals were analyzed. The oncology pharmacogene with the highest prevalence of high-risk, actionable genotypes was UGT1A1, impacting 17% of genotyped individuals. Actionable TPMT and DPYD genotypes were found in 9% and 4% of patients, respectively. Overall, nearly one-third of patients genotyped across all 5 genes (161/525, 31%) had at least one actionable genotype. CONCLUSIONS These data suggest that germline pharmacogenomic testing for 5 key pharmacogenes could identify a substantial proportion of patients at risk with standard dosing, an estimated impact similar to that of somatic genomic profiling. LAY SUMMARY Differences in our genes may explain why some drugs work safely in certain individuals but can cause side effects in others. Pharmacogenomics is the study of how genetic variations affect an individual's response to medications. In this study, an evaluation was done for important genetic variations that can affect the tolerability of anticancer therapy. By analyzing the genetic results of >1500 patients, it was found that nearly one-third have genetic variations that could alter recommendations of what drug, or how much of, an anticancer therapy they should be given. Performing pharmacogenomic testing before prescribing could help to guide personalized oncology care.
Collapse
Affiliation(s)
- Natalie M Reizine
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois.,Center for Personalized Therapeutics, University of Chicago, Chicago, Illinois
| | - Keith Danahey
- Center for Personalized Therapeutics, University of Chicago, Chicago, Illinois.,Center for Research Informatics, University of Chicago, Chicago, Illinois
| | - Tien M Truong
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois.,Center for Personalized Therapeutics, University of Chicago, Chicago, Illinois
| | - David George
- Center for Personalized Therapeutics, University of Chicago, Chicago, Illinois.,Department of Pathology, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois
| | - Larry K House
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois.,Center for Personalized Therapeutics, University of Chicago, Chicago, Illinois
| | - Theodore G Karrison
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| | - Xander M R van Wijk
- Center for Personalized Therapeutics, University of Chicago, Chicago, Illinois.,Department of Pathology, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois.,Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Kiang-Teck J Yeo
- Center for Personalized Therapeutics, University of Chicago, Chicago, Illinois.,Department of Pathology, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois.,Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Mark J Ratain
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois.,Center for Personalized Therapeutics, University of Chicago, Chicago, Illinois.,Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Peter H O'Donnell
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, Illinois.,Center for Personalized Therapeutics, University of Chicago, Chicago, Illinois.,Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| |
Collapse
|
4
|
Escala-Garcia M, Canisius S, Keeman R, Beesley J, Anton-Culver H, Arndt V, Augustinsson A, Becher H, Beckmann MW, Behrens S, Bermisheva M, Bojesen SE, Bolla MK, Brenner H, Canzian F, Castelao JE, Chang-Claude J, Chanock SJ, Couch FJ, Czene K, Daly MB, Dennis J, Devilee P, Dörk T, Dunning AM, Easton DF, Ekici AB, Eliassen AH, Fasching PA, Flyger H, Gago-Dominguez M, García-Closas M, García-Sáenz JA, Geisler J, Giles GG, Grip M, Gündert M, Hahnen E, Haiman CA, Håkansson N, Hall P, Hamann U, Hartikainen JM, Heemskerk-Gerritsen BAM, Hollestelle A, Hoppe R, Hopper JL, Hunter DJ, Jacot W, Jakubowska A, John EM, Jung AY, Kaaks R, Khusnutdinova E, Koppert LB, Kraft P, Kristensen VN, Kurian AW, Lambrechts D, Le Marchand L, Lindblom A, Luben RN, Lubiński J, Mannermaa A, Manoochehri M, Margolin S, Mavroudis D, Muranen TA, Nevanlinna H, Olshan AF, Olsson H, Park-Simon TW, Patel AV, Peterlongo P, Pharoah PDP, Punie K, Radice P, Rennert G, Rennert HS, Romero A, Roylance R, Rüdiger T, Ruebner M, Saloustros E, Sawyer EJ, Schmutzler RK, Schoemaker MJ, Scott C, Southey MC, Surowy H, Swerdlow AJ, Tamimi RM, Teras LR, Thomas E, Tomlinson I, Troester MA, Vachon CM, Wang Q, Winqvist R, Wolk A, et alEscala-Garcia M, Canisius S, Keeman R, Beesley J, Anton-Culver H, Arndt V, Augustinsson A, Becher H, Beckmann MW, Behrens S, Bermisheva M, Bojesen SE, Bolla MK, Brenner H, Canzian F, Castelao JE, Chang-Claude J, Chanock SJ, Couch FJ, Czene K, Daly MB, Dennis J, Devilee P, Dörk T, Dunning AM, Easton DF, Ekici AB, Eliassen AH, Fasching PA, Flyger H, Gago-Dominguez M, García-Closas M, García-Sáenz JA, Geisler J, Giles GG, Grip M, Gündert M, Hahnen E, Haiman CA, Håkansson N, Hall P, Hamann U, Hartikainen JM, Heemskerk-Gerritsen BAM, Hollestelle A, Hoppe R, Hopper JL, Hunter DJ, Jacot W, Jakubowska A, John EM, Jung AY, Kaaks R, Khusnutdinova E, Koppert LB, Kraft P, Kristensen VN, Kurian AW, Lambrechts D, Le Marchand L, Lindblom A, Luben RN, Lubiński J, Mannermaa A, Manoochehri M, Margolin S, Mavroudis D, Muranen TA, Nevanlinna H, Olshan AF, Olsson H, Park-Simon TW, Patel AV, Peterlongo P, Pharoah PDP, Punie K, Radice P, Rennert G, Rennert HS, Romero A, Roylance R, Rüdiger T, Ruebner M, Saloustros E, Sawyer EJ, Schmutzler RK, Schoemaker MJ, Scott C, Southey MC, Surowy H, Swerdlow AJ, Tamimi RM, Teras LR, Thomas E, Tomlinson I, Troester MA, Vachon CM, Wang Q, Winqvist R, Wolk A, Ziogas A, Michailidou K, Chenevix-Trench G, Bachelot T, Schmidt MK. Germline variants and breast cancer survival in patients with distant metastases at primary breast cancer diagnosis. Sci Rep 2021; 11:19787. [PMID: 34611289 PMCID: PMC8492709 DOI: 10.1038/s41598-021-99409-3] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 09/07/2021] [Indexed: 02/02/2023] Open
Abstract
Breast cancer metastasis accounts for most of the deaths from breast cancer. Identification of germline variants associated with survival in aggressive types of breast cancer may inform understanding of breast cancer progression and assist treatment. In this analysis, we studied the associations between germline variants and breast cancer survival for patients with distant metastases at primary breast cancer diagnosis. We used data from the Breast Cancer Association Consortium (BCAC) including 1062 women of European ancestry with metastatic breast cancer, 606 of whom died of breast cancer. We identified two germline variants on chromosome 1, rs138569520 and rs146023652, significantly associated with breast cancer-specific survival (P = 3.19 × 10-8 and 4.42 × 10-8). In silico analysis suggested a potential regulatory effect of the variants on the nearby target genes SDE2 and H3F3A. However, the variants showed no evidence of association in a smaller replication dataset. The validation dataset was obtained from the SNPs to Risk of Metastasis (StoRM) study and included 293 patients with metastatic primary breast cancer at diagnosis. Ultimately, larger replication studies are needed to confirm the identified associations.
Collapse
Affiliation(s)
- Maria Escala-Garcia
- Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Sander Canisius
- Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Renske Keeman
- Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Jonathan Beesley
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Hoda Anton-Culver
- Department of Medicine, Genetic Epidemiology Research Institute, University of California Irvine, Irvine, CA, USA
| | - Volker Arndt
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Annelie Augustinsson
- Department of Cancer Epidemiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Heiko Becher
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Sabine Behrens
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marina Bermisheva
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia
| | - Stig E Bojesen
- Copenhagen University Hospital, Copenhagen General Population Study, Herlev, Denmark
- Gentofte Hospital, Herlev, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Manjeet K Bolla
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jose E Castelao
- Instituto de Investigación Sanitaria Galicia Sur (IISGS), Xerencia de Xestion Integrada de Vigo-SERGAS, Oncology and Genetics Unit, Vigo, Spain
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- University Medical Center Hamburg-Eppendorf, Cancer Epidemiology Group, University Cancer Center Hamburg (UCCH), Hamburg, Germany
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mary B Daly
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Joe Dennis
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Peter Devilee
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Alison M Dunning
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Arif B Ekici
- Institute of Human Genetics, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Henrik Flyger
- Department of Breast Surgery, Copenhagen University Hospital, Herlev, Denmark
| | - Manuela Gago-Dominguez
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago, SERGAS, Fundación Pública Galega de Medicina Xenómica, Santiago de Compostela, Spain
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Montserrat García-Closas
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - José A García-Sáenz
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Centro Investigación Biomédica en Red de Cáncer (CIBERONC), Medical Oncology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | - Graham G Giles
- Cancer Council Victoria, Cancer Epidemiology Division, Melbourne, VIC, Australia
- Melbourne School of Population and Global Health, Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Mervi Grip
- Department of Surgery, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Melanie Gündert
- German Cancer Research Center (DKFZ), Molecular Epidemiology Group, C080, Heidelberg, Germany
- Molecular Biology of Breast Cancer, University Womens Clinic Heidelberg, University of Heidelberg, Heidelberg, Germany
- Helmholtz Zentrum München, Institute of Diabetes Research, German Research Center for Environmental Health, Neuherberg, Germany
| | - Eric Hahnen
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology (CIO), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Niclas Håkansson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Sšdersjukhuset, Stockholm, Sweden
| | - Ute Hamann
- German Cancer Research Center (DKFZ), Molecular Genetics of Breast Cancer, Heidelberg, Germany
| | - Jaana M Hartikainen
- Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
| | | | | | - Reiner Hoppe
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - John L Hopper
- Melbourne School of Population and Global Health, Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC, Australia
| | - David J Hunter
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - William Jacot
- Institut du Cancer de Montpellier, Montpellier University, Montpellier, France
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
- Independent Laboratory of Molecular Biology and Genetic Diagnostics, Pomeranian Medical University, Szczecin, Poland
| | - Esther M John
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford Cancer Institute, Stanford, CA, USA
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Audrey Y Jung
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elza Khusnutdinova
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia
- Department of Genetics and Fundamental Medicine, Bashkir State University, Ufa, Russia
| | - Linetta B Koppert
- Department of Surgical Oncology, Family Cancer Clinic, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Program in Genetic Epidemiology and Statistical Genetics, Boston, MA, USA
| | - Vessela N Kristensen
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Allison W Kurian
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford Cancer Institute, Stanford, CA, USA
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Diether Lambrechts
- VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, University of Leuven, Leuven, Belgium
| | - Loic Le Marchand
- University of Hawaii Cancer Center, Epidemiology Program, Honolulu, HI, USA
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Robert N Luben
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, England, UK
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Jan Lubiński
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Arto Mannermaa
- Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Kuopio University Hospital, Biobank of Eastern Finland, Kuopio, Finland
| | - Mehdi Manoochehri
- German Cancer Research Center (DKFZ), Molecular Genetics of Breast Cancer, Heidelberg, Germany
| | - Sara Margolin
- Department of Oncology, Sšdersjukhuset, Stockholm, Sweden
- Department of Clinical Science and Education, Karolinska Institutet, Sšdersjukhuset, Stockholm, Sweden
| | - Dimitrios Mavroudis
- Department of Medical Oncology, University Hospital of Heraklion, Heraklion, Greece
| | - Taru A Muranen
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Andrew F Olshan
- Department of Epidemiology, Gillings School of Global Public Health and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Håkan Olsson
- Department of Cancer Epidemiology, Clinical Sciences, Lund University, Lund, Sweden
| | | | - Alpa V Patel
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Paolo Peterlongo
- IFOM-The FIRC Institute of Molecular Oncology, Genome Diagnostics Program, Milan, Italy
| | - Paul D P Pharoah
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Paolo Radice
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori (INT), Milan, Italy
| | - Gad Rennert
- Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - Hedy S Rennert
- Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - Atocha Romero
- Medical Oncology Department, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | | | - Thomas Rüdiger
- Institute of Pathology, Staedtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - Matthias Ruebner
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | | | - Elinor J Sawyer
- School of Cancer & Pharmaceutical Sciences, Comprehensive Cancer Centre, King's College London, Guy's Campus, London, UK
| | - Rita K Schmutzler
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology (CIO), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Minouk J Schoemaker
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Christopher Scott
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Melissa C Southey
- Cancer Council Victoria, Cancer Epidemiology Division, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Harald Surowy
- German Cancer Research Center (DKFZ), Molecular Epidemiology Group, C080, Heidelberg, Germany
- Molecular Biology of Breast Cancer, University Womens Clinic Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Division of Breast Cancer Research, The Institute of Cancer Research, London, UK
| | - Rulla M Tamimi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Lauren R Teras
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Emilie Thomas
- Plateforme de Bioinformatique Gilles Thomas, Centre de recherche en cancérologie de Lyon, Fondation Synergie Lyon Cancer, Université Claude Bernard Lyon 1, Lyon, France
| | - Ian Tomlinson
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Wellcome Trust Centre for Human Genetics and Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Melissa A Troester
- Department of Epidemiology, Gillings School of Global Public Health and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Celine M Vachon
- Division of Epidemiology, Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Qin Wang
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Robert Winqvist
- Laboratory of Cancer Genetics and Tumor Biology, Cancer and Translational Medicine Research Unit, University of Oulu, Biocenter Oulu, Oulu, Finland
- Laboratory of Cancer Genetics and Tumor Biology, Northern Finland Laboratory Centre Oulu, Oulu, Finland
| | - Alicja Wolk
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Argyrios Ziogas
- Department of Medicine, Genetic Epidemiology Research Institute, University of California Irvine, Irvine, CA, USA
| | - Kyriaki Michailidou
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- Biostatistics Unit, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Georgia Chenevix-Trench
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Thomas Bachelot
- Département de Cancérologie Médicale, Centre Léon Bérard, Lyon, France
| | - Marjanka K Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Welm BE, Vaklavas C, Welm AL. Toward improved models of human cancer. APL Bioeng 2021; 5:010901. [PMID: 33415312 PMCID: PMC7785323 DOI: 10.1063/5.0030534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/30/2020] [Indexed: 12/27/2022] Open
Abstract
Human cancer is a complex and heterogeneous collection of diseases that kills
more than 18 million people every year worldwide. Despite advances in detection,
diagnosis, and treatments for cancers, new strategies are needed to combat
deadly cancers. Models of human cancer continue to evolve for preclinical
research and have culminated in patient-derived systems that better represent
the diversity and complexity of cancer. Still, no model is perfect. This
Perspective attempts to address ways that we can improve the clinical
translatability of models used for cancer research, from the point of view of
researchers who mainly conduct cancer studies in vivo.
Collapse
Affiliation(s)
- Bryan E Welm
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Christos Vaklavas
- Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
6
|
Reizine N, Vokes EE, Liu P, Truong TM, Nanda R, Fleming GF, Catenacci DV, Pearson AT, Parsad S, Danahey K, van Wijk XMR, Yeo KTJ, Ratain MJ, O’Donnell PH. Implementation of pharmacogenomic testing in oncology care (PhOCus): study protocol of a pragmatic, randomized clinical trial. Ther Adv Med Oncol 2020; 12:1758835920974118. [PMID: 33414846 PMCID: PMC7750903 DOI: 10.1177/1758835920974118] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/23/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Many cancer patients who receive chemotherapy experience adverse drug effects. Pharmacogenomics (PGx) has promise to personalize chemotherapy drug dosing to maximize efficacy and safety. Fluoropyrimidines and irinotecan have well-known germline PGx associations. At our institution, we have delivered PGx clinical decision support (CDS) based on preemptively obtained genotyping results for a large number of non-oncology medications since 2012, but have not previously evaluated the utility of this strategy for patients initiating anti-cancer regimens. We hypothesize that providing oncologists with preemptive germline PGx information along with CDS will enable individualized dosing decisions and result in improved patient outcomes. METHODS Patients with oncologic malignancies for whom fluoropyrimidine and/or irinotecan-inclusive therapy is being planned will be enrolled and randomly assigned to PGx and control arms. Patients will be genotyped in a clinical laboratory across panels that include actionable variants in UGT1A1 and DPYD. For PGx arm patients, treating providers will be given access to the patient-specific PGx results with CDS prior to treatment initiation. In the control arm, genotyping will be deferred, and dosing will occur as per usual care. Co-primary endpoints are dose intensity deviation rate (the proportion of patients receiving dose modifications during the first treatment cycle), and grade ⩾3 treatment-related toxicities throughout the treatment course. Additional study endpoints will include cumulative drug dose intensity, progression-free survival, dosing of additional PGx supportive medications, and patient-reported quality of life and understanding of PGx. DISCUSSION Providing a platform of integrated germline PGx information may promote personalized chemotherapy dosing decisions and establish a new model of care to optimize oncology treatment planning.
Collapse
Affiliation(s)
- Natalie Reizine
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, IL, USA
- Center for Personalized Therapeutics, University of Chicago, Chicago, IL, USA
| | - Everett E. Vokes
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, IL, USA
| | - Ping Liu
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Tien M. Truong
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, IL, USA
- Center for Personalized Therapeutics, University of Chicago, Chicago, IL, USA
| | - Rita Nanda
- Department of Pharmacy, University of Chicago Medical Center, Chicago, IL, USA
| | - Gini F. Fleming
- Department of Pharmacy, University of Chicago Medical Center, Chicago, IL, USA
| | | | | | - Sandeep Parsad
- Department of Pharmacy, University of Chicago Medical Center, Chicago, IL, USA
| | - Keith Danahey
- Center for Personalized Therapeutics, University of Chicago, Chicago, IL, USA Center for Research Informatics, University of Chicago, Chicago, IL, USA
| | - Xander M. R. van Wijk
- Center for Personalized Therapeutics, University of Chicago, Chicago, IL, USA Department of Pathology, University of Chicago Medical Center and Biological Sciences, Chicago, IL, USA
| | - Kiang-Teck J. Yeo
- Center for Personalized Therapeutics, University of Chicago, Chicago, IL, USA Department of Pathology, University of Chicago Medical Center and Biological Sciences, Chicago, IL, USA
| | - Mark J. Ratain
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, IL, USA Center for Personalized Therapeutics, University of Chicago, Chicago, IL, USA
| | - Peter H. O’Donnell
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center and Biological Sciences, Chicago, 5841 S. Maryland Avenue, MC2115, Chicago, IL 60637, USA
- Center for Personalized Therapeutics, University of Chicago, 5841 S. Maryland Avenue, MC2115, Chicago, IL 60637, USA
| |
Collapse
|
7
|
Caan BJ, Meyerhardt JA, Brown JC, Campbell KL, Cespedes Feliciano EM, Lee C, Ross MC, Quinney S, Quesenberry C, Sternfeld B, Schmitz KH. Recruitment strategies and design considerations in a trial of resistance training to prevent dose-limiting toxicities in colon cancer patients undergoing chemotherapy. Contemp Clin Trials 2020; 101:106242. [PMID: 33301991 DOI: 10.1016/j.cct.2020.106242] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/25/2020] [Accepted: 12/04/2020] [Indexed: 12/29/2022]
Abstract
Low muscle is associated with an increased risk of chemotherapy-related dose limiting toxicities (DLT) in cancer patients. Resistance training (RT) improves muscle mass; however, the effects of RT on preventing DLTs and dose reductions in colon cancer patients has not been investigated. FOcus on Reducing dose-limiting toxicities in Colon cancer with resistance Exercise (FORCE) is a multicenter, randomized clinical trial examining the effects of RT on relative dose intensity (RDI; primary outcome) and moderate and severe chemotoxicities (primary outcome) in non-metastatic colon cancer patients receiving adjuvant chemotherapy. Patients (N = 180) will be recruited from Kaiser Permanente Northern California, Dana-Farber Cancer Institute, and Penn State Cancer Institute. This paper describes recruitment strategies and design considerations. Patients will be randomized in equal numbers to RT intervention or control. Patients have baseline and post completion of chemotherapy visits where information on anthropometry, physical function, body composition, quality of life, physical activity and dietary behaviors, and inflammatory blood markers will be collected. Patient-reported outcomes of chemotherapy side effects will be collected around the time of chemotherapy throughout the duration of the trial. Intervention participants will be prescribed a progressive RT program consisting of 4-6 visits with a certified exercise trainer, delivered either in-person or remotely by video conference, and will be asked to engage twice weekly in-home training sessions. Control patients at the end of the study receive a consult with a FORCE exercise trainer, an online exercise RT training program and a set of resistance bands. Results of this trial will provide information on the benefit of resistance exercise as a treatment to increase RDI.
Collapse
Affiliation(s)
- Bette J Caan
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA.
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA 02215, USA.
| | - Justin C Brown
- Cancer Metabolism Research Program, Pennington Biomedical Research Center, Louisiana State University, 6400 Perkins Road, Baton Rouge, Louisiana 70808, USA.
| | - Kristin L Campbell
- Department of Physical Therapy, University of British Columbia, 212 Friedman Building, 2177 Westbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada.
| | | | - Catherine Lee
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA.
| | - Michelle C Ross
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA.
| | - Sara Quinney
- Disease and Therapeutic Response Modeling Program, Indiana University School of Medicine, 950 W Walnut Street, Indianapolis, Indiana 46202, USA.
| | - Charles Quesenberry
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA.
| | - Barbara Sternfeld
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
| | - Kathryn H Schmitz
- Penn State Cancer Institute, The Pennsylvania State University, 500 University Drive, Hershey, PA 17033, USA.
| |
Collapse
|
8
|
Ryden LE, Lewis SJG. Parkinson's Disease in the Era of Personalised Medicine: One Size Does Not Fit All. Drugs Aging 2019; 36:103-113. [PMID: 30556112 DOI: 10.1007/s40266-018-0624-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The concept of personalised medicine in Parkinson's disease has arrived where the implications of findings made in research are certain to have an increasing impact upon clinical practice. Disease heterogeneity in Parkinson's disease has been well described and lends itself to the construct of personalised medicine where it is hypothesised that a greater understanding of genetic and pathophysiological contributions may underpin the sub-groups described. This in turn has driven the development of potentially individualised disease-modifying therapies where, for example, we are beginning to see treatments that target patients with Parkinson's disease with specific genetic mutations. Furthermore, clinicians are increasingly recognising the need to tailor their management approach to patients depending on their age of presentation, acknowledging differential side-effect profiles and responses especially when considering the use of device-assisted technologies such as infusion or surgery. Clearly, individualising the treatment of both motor and non-motor symptoms will remain imperative but, in the future, personalised medicine may provide clearer insights into various aspects of a patient's symptomatology, disease course and thus the best therapeutic approaches.
Collapse
Affiliation(s)
- Lauren E Ryden
- Parkinson's Disease Research Clinic, Brain and Mind Centre, University of Sydney, 100 Mallett St, Camperdown, NSW, 2050, Australia
| | - Simon J G Lewis
- Parkinson's Disease Research Clinic, Brain and Mind Centre, University of Sydney, 100 Mallett St, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
9
|
Giannopoulou E, Katsila T, Mitropoulou C, Tsermpini EE, Patrinos GP. Integrating Next-Generation Sequencing in the Clinical Pharmacogenomics Workflow. Front Pharmacol 2019; 10:384. [PMID: 31024324 PMCID: PMC6460422 DOI: 10.3389/fphar.2019.00384] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/27/2019] [Indexed: 12/12/2022] Open
Abstract
Pharmacogenomics has been recognized as a fundamental tool in the era of personalized medicine with up to 266 drug labels, approved by major regulatory bodies, currently containing pharmacogenomics information. Next-generation sequencing analysis assumes a critical role in personalized medicine, providing a comprehensive profile of an individual's variome, particularly that of clinical relevance, comprising of pathogenic variants and pharmacogenomic biomarkers. Here, we propose a strategy to integrate next-generation sequencing into the current clinical pharmacogenomics workflow from deep resequencing to pharmacogenomics consultation, according to the existing guidelines and recommendations.
Collapse
Affiliation(s)
| | - Theodora Katsila
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece
| | | | | | - George P Patrinos
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece.,Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.,Zayed Center of Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| |
Collapse
|
10
|
Impact of Occupational Exposures and Genetic Polymorphisms on Recurrence and Progression of Non-Muscle-Invasive Bladder Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15081563. [PMID: 30042310 PMCID: PMC6121504 DOI: 10.3390/ijerph15081563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/10/2018] [Accepted: 07/20/2018] [Indexed: 12/13/2022]
Abstract
Introduction: Additional or better markers are needed to guide the clinical monitoring of patients with non-muscle-invasive bladder cancer (NMIBC). Aim: To investigate the influence of occupational exposures and genetic polymorphisms on recurrence and progression of NMIBC. Methods: The study includes 160 NMIBC patients. We collected on questionnaire information on demographic variables, lifetime smoking history, lifetime history of occupational exposure to aromatic amines and polycyclic aromatic hydrocarbons. Genetic polymorphism (glutathione S-transferase M1; T1; P1 (GSTM1; GSTT1; GSTP1); N-acetyltransferase 1; 2 (NAT1; NAT2); cytochrome P450 1B1 (CYP1B1); sulfotransferase 1A1 (SULT1A1); myeloperoxidase (MPO); catechol-O-methyltransferase (COMT); manganese superoxide dismutase (MnSOD); NAD(P)H:quinone oxidoreductase (NQO1); X-ray repair cross-complementing group 1; 3 (XRCC1; XRCC3) and xeroderma pigmentosum complementation group (XPD)) was assessed in peripheral blood lymphocytes. DNA adducts were evaluated by 32P-postlabeling. Predictors of recurrence (histological confirmation of a newly found bladder tumor) and progression (transition of tumor from low-grade to high-grade and/or increase in TNM stage) were identified by multivariate Cox proportional hazard regression with stepwise backward selection of independent variables. Hazard ratios (HR) with 95% confidence interval (95%CI) and two-tail probability of error (p-value) were estimated. Results: The risk of BC progression decreased with the homozygous genotype “ValVal” of both COMT and MnSOD (HR = 0.195; 95%CI = 0.060 to 0.623; p = 0.006). The results on BC recurrence were of borderline significance. No occupational exposure influenced recurrence or progression. Conclusion: Our results are supported by experimental evidence of a plausible mechanism between cause (ValVal genotype of both MnSOD and COMT) and effect (decreased progression of tumor in NMIBC patients). The genetic polymorphisms associated with better prognosis may be used in clinic to guide selection of treatment for patients initially diagnosed with NMIBC. However, external validation studies are required.
Collapse
|
11
|
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
|
12
|
Abstract
Future medical technology breakthroughs will build from the incredible progress made in computers, biotechnology, and nanotechnology and from the information learned from the human genome. With such technology and information, computer-aided diagnoses, organ replacement, gene therapy, personalized drugs, and even age reversal will become possible. True 3-dimensional system technology will enable surgeons to envision key clinical features and will help them in planning complex surgery. Surgeons will enter surgical instructions in a virtual space from a remote medical center, order a medical robot to perform the operation, and review the operation in real time on a monitor. Surgeons will be better than artificial intelligence or automated robots when surgeons (or we) love patients and ask questions for a better future. The purpose of this paper is looking at the future medical science and the changes of colorectal surgeons.
Collapse
Affiliation(s)
- Young Jin Kim
- Department of Surgery, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
13
|
Dias MM, Sorich MJ, Rowland A, Wiese MD, McKinnon RA. The Routine Clinical use of Pharmacogenetic Tests: What it Will Require? Pharm Res 2017; 34:1544-1550. [PMID: 28236061 DOI: 10.1007/s11095-017-2128-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/16/2017] [Indexed: 01/04/2023]
Abstract
Pharmacogenetic testing aims to personalize drug therapy with a view to optimising drug efficacy and minimise toxicity. However, despite the potential benefits, pharmacogenetic testing is mostly confined to specialised medical areas, laboratories and centres. Widespread integration into routine clinical practice has been limited by a complex set of issues including regulatory and reimbursement frameworks, evidence of clinical utility and clinician perspectives, practices and education. Here we assess the current barriers to widespread clinical uptake and identify the key issue necessary to address to accelerate routine testing.
Collapse
Affiliation(s)
- Mafalda M Dias
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia.,Department of Clinical Pharmacology, School of Medicine, Flinders University, Bedford Park, Australia
| | - Michael J Sorich
- Department of Clinical Pharmacology, School of Medicine, Flinders University, Bedford Park, Australia.,Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, GPO Box 2100, Adelaide, SA, 5001, Australia
| | - Andrew Rowland
- Department of Clinical Pharmacology, School of Medicine, Flinders University, Bedford Park, Australia.,Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, GPO Box 2100, Adelaide, SA, 5001, Australia
| | - Michael D Wiese
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Ross A McKinnon
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, GPO Box 2100, Adelaide, SA, 5001, Australia.
| |
Collapse
|
14
|
Lynch JA, Berse B, Dotson WD, Khoury MJ, Coomer N, Kautter J. Utilization of genetic tests: analysis of gene-specific billing in Medicare claims data. Genet Med 2017; 19:890-899. [PMID: 28125086 DOI: 10.1038/gim.2016.209] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/17/2016] [Indexed: 02/06/2023] Open
Abstract
PURPOSE We examined the utilization of precision medicine tests among Medicare beneficiaries through analysis of gene-specific tier 1 and 2 billing codes developed by the American Medical Association in 2012. METHODS We conducted a retrospective cross-sectional study. The primary source of data was 2013 Medicare 100% fee-for-service claims. We identified claims billed for each laboratory test, the number of patients tested, expenditures, and the diagnostic codes indicated for testing. We analyzed variations in testing by patient demographics and region of the country. RESULTS Pharmacogenetic tests were billed most frequently, accounting for 48% of the expenditures for new codes. The most common indications for testing were breast cancer, long-term use of medications, and disorders of lipid metabolism. There was underutilization of guideline-recommended tumor mutation tests (e.g., epidermal growth factor receptor) and substantial overutilization of a test discouraged by guidelines (methylenetetrahydrofolate reductase). Methodology-based tier 2 codes represented 15% of all claims billed with the new codes. The highest rate of testing per beneficiary was in Mississippi and the lowest rate was in Alaska. CONCLUSIONS Gene-specific billing codes significantly improved our ability to conduct population-level research of precision medicine. Analysis of these data in conjunction with clinical records should be conducted to validate findings.Genet Med advance online publication 26 January 2017.
Collapse
Affiliation(s)
- Julie A Lynch
- RTI International, Waltham, Massachusetts, USA.,Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA
| | - Brygida Berse
- RTI International, Waltham, Massachusetts, USA.,Boston University School of Medicine, Boston, Massachusetts, USA.,Veterans Health Administration, Bedford, Massachusetts, USA
| | - W David Dotson
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Muin J Khoury
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nicole Coomer
- RTI International, Research Triangle Park, North Carolina, USA
| | | |
Collapse
|
15
|
Hirschtritt ME, Besterman AD, Ross DA. Psychiatric Pharmacogenomics: How Close Are We? Biol Psychiatry 2016; 80:e63-5. [PMID: 27663067 PMCID: PMC5371713 DOI: 10.1016/j.biopsych.2016.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 11/29/2022]
Affiliation(s)
| | - Aaron D Besterman
- Department of Psychiatry, University of California Los Angeles, Los Angeles, California
| | - David A Ross
- Department of Psychiatry, Yale University, New Haven, Connecticut.
| |
Collapse
|
16
|
O'Donnell PH, Alanee S, Stratton KL, Garcia-Grossman IR, Cao H, Ostrovnaya I, Plimack ER, Manschreck C, Ganshert C, Smith ND, Steinberg GD, Vijai J, Offit K, Stadler WM, Bajorin DF. Clinical Evaluation of Cisplatin Sensitivity of Germline Polymorphisms in Neoadjuvant Chemotherapy for Urothelial Cancer. Clin Genitourin Cancer 2016; 14:511-517. [PMID: 27150640 DOI: 10.1016/j.clgc.2016.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/03/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND Level 1 evidence has demonstrated increased overall survival with cisplatin-based neoadjuvant chemotherapy for patients with muscle-invasive urothelial cancer. Usage remains low, however, in part because neoadjuvant chemotherapy will not be effective for every patient. To identify the patients most likely to benefit, we evaluated germline pharmacogenomic markers for association with neoadjuvant chemotherapy sensitivity in 2 large cohorts of patients with urothelial cancer. PATIENTS AND METHODS Patients receiving neoadjuvant cisplatin-based chemotherapy for muscle-invasive urothelial cancer were eligible. Nine germline single nucleotide polymorphisms (SNPs) potentially conferring platinum sensitivity were tested for an association with a complete pathologic response to neoadjuvant chemotherapy (pT0) or elimination of muscle-invasive cancer (<pT2). RESULTS The data from 205 patients were analyzed-59 patients were included in the discovery set and 146 in an independent replication cohort-from 3 institutions. The stage pT0 (26%) and < pT2 (50%) rates were consistent across the discovery and replication populations. Using a multivariate recessive genetic model, rs244898 in RARS (odds ratio, 6.8; 95% confidence interval, 1.8-28.9; P = .006) and rs7937567 in GALNTL4 (odds ratio, 4.8; 95% confidence interval, 1.1-22.6; P = .04) were associated with pT0 in the discovery set. Despite these large effects, neither were associated with achievement of pT0 in the replication set. A third SNP, rs10964552, was associated with stage < pT2 in the discovery set but also failed to replicate. CONCLUSION Germline SNPs previously associated with platinum sensitivity were not associated with the neoadjuvant chemotherapy response in a large replication cohort of patients with urothelial cancer. These results emphasize the need for replication when evaluating pharmacogenomic markers and demonstrate that multi-institutional efforts are feasible and will be necessary to achieve advances in urothelial cancer pharmacogenomics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Joseph Vijai
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kenneth Offit
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | |
Collapse
|
17
|
Ruwali M, Dhawan A, Pant MC, Rahman Q, Khurana SP, Parmar D. Clinical Management of Head and Neck Cancer Cases: Role of Pharmacogenetics of CYP2 and GSTs. Oncol Res Treat 2016; 39:221-6. [DOI: 10.1159/000444608] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 02/05/2016] [Indexed: 11/19/2022]
|
18
|
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]
|
19
|
Hattinger CM, Serra M. Role of pharmacogenetics of drug-metabolizing enzymes in treating osteosarcoma. Expert Opin Drug Metab Toxicol 2015; 11:1449-63. [PMID: 26095223 DOI: 10.1517/17425255.2015.1060220] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Drug-metabolizing enzymes (DMEs) biotransform several toxins and xenobiotics in both tumor and normal cells, resulting in either their detoxification or their activation. Since DMEs also metabolize several chemotherapeutic drugs, they can significantly influence tumor response to chemotherapy and susceptibility of normal tissues to collateral toxicity of anticancer treatments. AREAS COVERED This review discusses the pharmacogenetics of DMEs involved in the metabolism of drugs which constitute the backbone of osteosarcoma (OS) chemotherapy, highlighting what is presently known for this tumor and their possible impact on the modulation of future treatment approaches. EXPERT OPINION Achieving further insight into pharmacogenetic markers and biological determinants related to treatment response in OS may ultimately lead to individualized treatment regimens, based on a combination of genotype and tumor characteristics of each patient.
Collapse
Affiliation(s)
- Claudia Maria Hattinger
- a Orthopaedic Rizzoli Institute, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit , Via di Barbiano 1/10, I-40136 Bologna, Italy +390 516 366 762 ; +390 516 366 763 ;
| | | |
Collapse
|
20
|
Kummar S, Williams PM, Lih CJ, Polley EC, Chen AP, Rubinstein LV, Zhao Y, Simon RM, Conley BA, Doroshow JH. Application of molecular profiling in clinical trials for advanced metastatic cancers. J Natl Cancer Inst 2015; 107:djv003. [PMID: 25663694 DOI: 10.1093/jnci/djv003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is growing interest in the application of molecular profiling, including sequencing, genotyping, and/or mRNA expression profiling, to the analysis of patient tumors with the objective of applying these data to inform therapeutic choices for patients with advanced cancers. Multiple clinical trials that are attempting to validate this personalized or precision medicine approach are in various stages of development and execution. Although preliminary data from some of these efforts have fueled excitement about the value and utility of these studies, their execution has also provoked many questions about the best way to approach complicating factors such as tumor heterogeneity and the choice of which genetic mutations to target. This commentary highlights some of the challenges confronting the clinical application of molecular tumor profiling and the various trial designs being utilized to address these challenges. Randomized trials that rigorously test patient response to molecularly targeted agents assigned based on the presence of a defined set of mutations in putative cancer-driving pathways are required to address some of the current challenges and to identify patients likely to benefit from this approach.
Collapse
Affiliation(s)
- Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (SK, ECP, APC, LVR, YZ, RMS, BAC, JHD); Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD (PMW, CJL).
| | - P Mickey Williams
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (SK, ECP, APC, LVR, YZ, RMS, BAC, JHD); Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD (PMW, CJL)
| | - Chih-Jian Lih
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (SK, ECP, APC, LVR, YZ, RMS, BAC, JHD); Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD (PMW, CJL)
| | - Eric C Polley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (SK, ECP, APC, LVR, YZ, RMS, BAC, JHD); Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD (PMW, CJL)
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (SK, ECP, APC, LVR, YZ, RMS, BAC, JHD); Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD (PMW, CJL)
| | - Larry V Rubinstein
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (SK, ECP, APC, LVR, YZ, RMS, BAC, JHD); Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD (PMW, CJL)
| | - Yingdong Zhao
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (SK, ECP, APC, LVR, YZ, RMS, BAC, JHD); Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD (PMW, CJL)
| | - Richard M Simon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (SK, ECP, APC, LVR, YZ, RMS, BAC, JHD); Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD (PMW, CJL)
| | - Barbara A Conley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (SK, ECP, APC, LVR, YZ, RMS, BAC, JHD); Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD (PMW, CJL)
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (SK, ECP, APC, LVR, YZ, RMS, BAC, JHD); Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD (PMW, CJL)
| |
Collapse
|
21
|
Campbell JM, Peters MDJ. The association of chemotherapy-induced toxicities with germline polymorphisms: an umbrella review of systematic reviews and meta-analyses. ACTA ACUST UNITED AC 2014. [DOI: 10.11124/jbisrir-2014-1877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
22
|
Gillis NK, Patel JN, Innocenti F. Clinical implementation of germ line cancer pharmacogenetic variants during the next-generation sequencing era. Clin Pharmacol Ther 2014; 95:269-80. [PMID: 24136381 PMCID: PMC4128332 DOI: 10.1038/clpt.2013.214] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 10/12/2013] [Indexed: 01/13/2023]
Abstract
More than 100 medications approved by the US Food and Drug Administration include pharmacogenetic biomarkers in the drug label, many with cancer indications referencing germ line DNA variations. With the advent of next-generation sequencing (NGS) and its rapidly increasing uptake into cancer research and clinical practice, an enormous amount of data to inform documented gene-drug associations will be collected that must be exploited to optimize patient benefit. This review focuses on the implementation of germ line cancer pharmacogenetics in clinical practice. Specifically, it discusses the importance of germ line variation in cancer and the role of NGS in pharmacogenetic discovery and implementation. In the context of a scenario in which massive amounts of NGS-based genetic information will be increasingly available to health stakeholders, this review explores the ongoing debate regarding the threshold of evidence necessary for implementation, provides an overview of recommendations in cancer by professional organizations and regulatory bodies, and discusses limitations of current guidelines and strategies to improve third-party coverage.
Collapse
Affiliation(s)
- Nancy K. Gillis
- Eshelman School of Pharmacy, Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC
| | - Jai N. Patel
- Eshelman School of Pharmacy, Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC
| | - Federico Innocenti
- Eshelman School of Pharmacy, Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
23
|
Grotenhuis AJ, Dudek AM, Verhaegh GW, Witjes JA, Aben KK, van der Marel SL, Vermeulen SH, Kiemeney LA. Prognostic relevance of urinary bladder cancer susceptibility loci. PLoS One 2014; 9:e89164. [PMID: 24586564 PMCID: PMC3934869 DOI: 10.1371/journal.pone.0089164] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 01/16/2014] [Indexed: 12/13/2022] Open
Abstract
In the last few years, susceptibility loci have been identified for urinary bladder cancer (UBC) through candidate-gene and genome-wide association studies. Prognostic relevance of most of these loci is yet unknown. In this study, we used data of the Nijmegen Bladder Cancer Study (NBCS) to perform a comprehensive evaluation of the prognostic relevance of all confirmed UBC susceptibility loci. Detailed clinical data concerning diagnosis, stage, treatment, and disease course of a population-based series of 1,602 UBC patients were collected retrospectively based on a medical file survey. Kaplan-Meier survival analyses and Cox proportional hazard regression were performed, and log-rank tests calculated, to evaluate the association between 12 confirmed UBC susceptibility variants and recurrence and progression in non-muscle invasive bladder cancer (NMIBC) patients. Among muscle-invasive or metastatic bladder cancer (MIBC) patients, association of these variants with overall survival was tested. Subgroup analyses by tumor aggressiveness and smoking status were performed in NMIBC patients. In the overall NMIBC group (n = 1,269), a statistically significant association between rs9642880 at 8q24 and risk of progression was observed (GT vs. TT: HR = 1.08 (95% CI: 0.76-1.54), GG vs. TT: HR = 1.81 (95% CI: 1.23-2.66), P for trend = 2.6 × 10(-3)). In subgroup analyses, several other variants showed suggestive, though non-significant, prognostic relevance for recurrence and progression in NMIBC and survival in MIBC. This study provides suggestive evidence that genetic loci involved in UBC etiology may influence disease prognosis. Elucidation of the causal variant(s) could further our understanding of the mechanism of disease, could point to new therapeutic targets, and might aid in improvement of prognostic tools.
Collapse
Affiliation(s)
- Anne J. Grotenhuis
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Aleksandra M. Dudek
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerald W. Verhaegh
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J. Alfred Witjes
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Katja K. Aben
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
- Comprehensive Cancer Center The Netherlands, Utrecht, The Netherlands
| | | | - Sita H. Vermeulen
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lambertus A. Kiemeney
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Integrating cell-based and clinical genome-wide studies to identify genetic variants contributing to treatment failure in neuroblastoma patients. Clin Pharmacol Ther 2014; 95:644-52. [PMID: 24549002 PMCID: PMC4029857 DOI: 10.1038/clpt.2014.37] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 02/11/2014] [Indexed: 12/02/2022]
Abstract
High-risk neuroblastoma is an aggressive malignancy with high rates of treatment failure. We evaluated genetic variants associated with in vitro sensitivity to two derivatives of cyclophosphamide for association with clinical response in a separate replication cohort of neuroblastoma patients (n=2,709). Lymphoblastoid cell lines (LCLs) were exposed to increasing concentrations of 4-hydroperoxycyclophosphamide [4HC n=422] and phosphoramide mustard [PM n=428] to determine sensitivity. Genome-wide association studies (GWAS) were performed to identify single nucleotide polymorphisms (SNPs) associated with 4HC and PM sensitivity. SNPs consistently associated with LCL sensitivity were analyzed for associations with event-free survival in patients. Two linked SNPs, rs9908694 and rs1453560, were found to be associated with PM sensitivity in LCLs across populations and were associated with event-free survival in all patients (P=0.01) and within the high-risk subset (P=0.05). Our study highlights the value of cell-based models to identify candidate variants that may predict response to treatment in patients with cancer.
Collapse
|
25
|
Zardavas D, Pugliano L, Piccart M. Personalized therapy for breast cancer: a dream or a reality? Future Oncol 2013; 9:1105-19. [PMID: 23902243 DOI: 10.2217/fon.13.57] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Breast cancer oncology represents one of the disciplines where personalized cancer medicine has been most actively pursued. The class-discovery studies conceptually advanced the field, underlining the molecular heterogeneity governing this common disease. The advent of high-throughput molecular profiling technologies holds great promise for the advance of all aspects of personalized cancer medicine, namely accurate prognostication, prediction of response to common systemic therapies and individualized monitoring of the disease. Moreover, an ever-expanding arsenal of targeted therapeutic compounds under clinical development, coupled with emerging powerful tools for comprehensive molecular and functional characterization, pose significant promise for improved clinical outcomes for breast cancer patients. Interrogation of the germline genetic variation offers further promise towards tailoring of breast cancer management. Well-conducted prospective validation studies are needed if breast cancer personalized therapy is to transform from a dream into a reality.
Collapse
Affiliation(s)
- Dimitrios Zardavas
- Institut Jules Bordet, Boulevard de Waterloo, 125, 1000 Brussels, Belgium
| | | | | |
Collapse
|
26
|
Monks NR, Cherba DM, Kamerling SG, Simpson H, Rusk AW, Carter D, Eugster E, Mooney M, Sigler R, Steensma M, Grabinski T, Marotti KR, Webb CP. A multi-site feasibility study for personalized medicine in canines with osteosarcoma. J Transl Med 2013; 11:158. [PMID: 23815880 PMCID: PMC3702405 DOI: 10.1186/1479-5876-11-158] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 06/28/2013] [Indexed: 01/06/2023] Open
Abstract
Background A successful therapeutic strategy, specifically tailored to the molecular constitution of an individual and their disease, is an ambitious objective of modern medicine. In this report, we highlight a feasibility study in canine osteosarcoma focused on refining the infrastructure and processes required for prospective clinical trials using a series of gene expression-based Personalized Medicine (PMed) algorithms to predict suitable therapies within 5 days of sample receipt. Methods Tumor tissue samples were collected immediately following limb amputation and shipped overnight from veterinary practices. Upon receipt (day 1), RNA was extracted from snap-frozen tissue, with an adjacent H&E section for pathological diagnosis. Samples passing RNA and pathology QC were shipped to a CLIA-certified laboratory for genomic profiling. After mapping of canine probe sets to human genes and normalization against a (normal) reference set, gene level Z-scores were submitted to the PMed algorithms. The resulting PMed report was immediately forwarded to the veterinarians. Upon receipt and review of the PMed report, feedback from the practicing veterinarians was captured. Results 20 subjects were enrolled over a 5 month period. Tissue from 13 subjects passed both histological and RNA QC and were submitted for genomic analysis and subsequent PMed analysis and report generation. 11 of the 13 samples for which PMed reports were produced were communicated to the veterinarian within the target 5 business days. Of the 7 samples that failed QC, 4 were due to poor RNA quality, whereas 2 were failed following pathological review. Comments from the practicing veterinarians were generally positive and constructive, highlighting a number of areas for improvement, including enhanced education regarding PMed report interpretation, drug availability, affordable pricing and suitable canine dosing. Conclusions This feasibility trial demonstrated that with the appropriate infrastructure and processes it is possible to perform an in-depth molecular analysis of a patient’s tumor in support of real time therapeutic decision making within 5 days of sample receipt. A number of areas for improvement have been identified that should reduce the level of sample attrition and support clinical decision making.
Collapse
|
27
|
O'Donnell PH. Bladder cancer pharmacogenomics: recent insights and future perspectives. Pharmacogenomics 2013; 13:1553-6. [PMID: 23148629 DOI: 10.2217/pgs.12.145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
28
|
Vianna-Jorge R, Festa-Vasconcellos JS, Goulart-Citrangulo SMT, Leite MS. Functional polymorphisms in xenobiotic metabolizing enzymes and their impact on the therapy of breast cancer. Front Genet 2013; 3:329. [PMID: 23346096 PMCID: PMC3551254 DOI: 10.3389/fgene.2012.00329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 12/29/2012] [Indexed: 01/21/2023] Open
Abstract
Breast cancer is the top cancer among women, and its incidence is increasing worldwide. Although the mortality tends to decrease due to early detection and treatment, there is great variability in the rates of clinical response and survival, which makes breast cancer one of the most appealing targets for pharmacogenomic studies. The recognition that functional CYP2D6 polymorphisms affect tamoxifen pharmacokinetics has motivated the attempts of using CYP2D6 genotyping for predicting breast cancer outcomes. In addition to tamoxifen, the chemotherapy of breast cancer includes combinations of cytotoxic drugs, which are substrates for various xenobiotic metabolizing enzymes. Because of these drugs’ narrow therapeutic window, it has been postulated that impaired biotransformation could lead to increased toxicity. In the present review, we performed a systematic search of all published data exploring associations between polymorphisms in xenobiotic metabolizing enzymes and clinical outcomes of breast cancer. We retrieved 43 original articles involving either tamoxifen or other chemotherapeutic protocols, and compiled all information regarding response or toxicity. The data indicate that, although CYP2D6 polymorphisms can indeed modify tamoxifen pharmacokinetics, CYP2D6 genotyping alone is not enough for predicting breast cancer outcomes. The studies involving other chemotherapeutic protocols explored a great diversity of pharmacogenetic targets, but the number of studies for each functional polymorphism is still very limited, with usually no confirmation of positive associations. In conclusion, the application of pharmacogenetics to predict breast cancer outcomes and to select one individual’s chemotherapeutic protocol is still far from clinical routine. Although some very interesting results have been produced, no clear practical recommendations are recognized yet.
Collapse
Affiliation(s)
- Rosane Vianna-Jorge
- Programa de Farmacologia, Coordenação de Pesquisa, Instituto Nacional do Câncer Rio de Janeiro, Brazil ; Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | | | | | | |
Collapse
|
29
|
Santos EMM, Edwards QT, Floria-Santos M, Rogatto SR, Achatz MIW, MacDonald DJ. Integration of Genomics in Cancer Care. J Nurs Scholarsh 2013; 45:43-51. [DOI: 10.1111/j.1547-5069.2012.01465.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Zafra-Ceres M, de Haro T, Farez-Vidal E, Blancas I, Bandres F, de Dueñas EM, Ochoa-Aranda E, Gomez-Capilla JA, Gomez-Llorente C. Influence of CYP2D6 polymorphisms on serum levels of tamoxifen metabolites in Spanish women with breast cancer. Int J Med Sci 2013; 10:932-7. [PMID: 23781139 PMCID: PMC3675507 DOI: 10.7150/ijms.5708] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 05/08/2013] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Estrogen receptor-positive breast cancer tumors depend on estrogen signaling for their growth and replication and can be treated by anti-estrogen therapy with tamoxifen. Polymorphisms of the CYP2D6 and CYP2C19 genes are associated with an impaired response to tamoxifen. The study objective was to investigate the impact of genetic polymorphisms in CYP2D6 and CYP2C19 on the pharmacokinetics of tamoxifen and its metabolites in Spanish women with estrogen receptor-positive breast cancer who were candidates for tamoxifen therapy. METHODS We studied 90 women with estrogen receptor-positive breast cancer, using the AmpliChip CYP450 test to determine CYP2D6 and CYP2C19 gene variants. Plasma levels of tamoxifen and its metabolites were quantified by high-performance liquid chromatography. RESULTS The CYP2D6 phenotype was extensive metabolizer in 80%, intermediate metabolizer in 12.2%, ultra-rapid metabolizer in 2.2%, and poor metabolizer in 5.6% of patients, and the allele frequency was 35.0% for allele (*)1, 21.0% for *2, and 18.9% for *4. All poor metabolizers in this series were *4/*4, and their endoxifen and 4-hydroxy tamoxifen levels were 25% lower than those of extensive metabolizers. CYP2C19*2 allele, which has been related to breast cancer outcomes, was detected in 15.6% of the studied alleles. CONCLUSION CYP2D6*4/*4 genotype was inversely associated with 4-hydroxy tamoxifen and endoxifen levels. According to these results, CYP2D6 and CYP2C19 genotyping appears advisable before the prescription of tamoxifen therapy.
Collapse
Affiliation(s)
- Mercedes Zafra-Ceres
- Clinical Biochemistry Services, San Cecilio University Hospital. Avd/ Doctor Olóriz s/n 18012, Granada, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Mechanisms of resistance to endocrine therapy in breast cancer: focus on signaling pathways, miRNAs and genetically based resistance. Int J Mol Sci 2012; 14:108-45. [PMID: 23344024 PMCID: PMC3565254 DOI: 10.3390/ijms14010108] [Citation(s) in RCA: 192] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/10/2012] [Accepted: 12/12/2012] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is the most frequent malignancy diagnosed in women. Approximately 70% of breast tumors express the estrogen receptor (ER). Tamoxifen and aromatase inhibitors (AIs) are the most common and effective therapies for patients with ERα-positive breast cancer. Alone or combined with chemotherapy, tamoxifen significantly reduces disease progression and is associated with more favorable impact on survival in patients. Unfortunately, endocrine resistance occurs, either de novo or acquired during the course of the treatment. The mechanisms that contribute to hormonal resistance include loss or modification in the ERα expression, regulation of signal transduction pathways, altered expression of specific microRNAs, balance of co-regulatory proteins, and genetic polymorphisms involved in tamoxifen metabolic activity. Because of the clinical consequences of endocrine resistance, new treatment strategies are arising to make the cells sensitive to tamoxifen. Here, we will review the current knowledge on mechanisms of endocrine resistance in breast cancer cells. In addition, we will discuss novel therapeutic strategies to overcome such resistance. Undoubtedly, circumventing endocrine resistance should help to improve therapy for the benefit of breast cancer patients.
Collapse
|
32
|
Moen EL, Godley LA, Zhang W, Dolan ME. Pharmacogenomics of chemotherapeutic susceptibility and toxicity. Genome Med 2012; 4:90. [PMID: 23199206 PMCID: PMC3580423 DOI: 10.1186/gm391] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The goal of personalized medicine is to tailor a patient's treatment strategy on the basis of his or her unique genetic make-up. The field of oncology is beginning to incorporate many of the strategies of personalized medicine, especially within the realm of pharmacogenomics, which is the study of how inter-individual genetic variation determines drug response or toxicity. A main objective of pharmacogenomics is to facilitate physician decision-making regarding optimal drug selection, dose and treatment duration on a patient-by-patient basis. Recent advances in genome-wide genotyping and sequencing technologies have supported the discoveries of a number of pharmacogenetic markers that predict response to chemotherapy. However, effectively implementing these pharmacogenetic markers in the clinic remains a major challenge. This review focuses on the contribution of germline genetic variation to chemotherapeutic toxicity and response, and discusses the utility of genome-wide association studies and use of lymphoblastoid cell lines (LCLs) in pharmacogenomic studies. Furthermore, we highlight several recent examples of genetic variants associated with chemotherapeutic toxicity or response in both patient cohorts and LCLs, and discuss the challenges and future directions of pharmacogenomic discovery for cancer treatment.
Collapse
Affiliation(s)
- Erika L Moen
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Lucy A Godley
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
- The University of Chicago Comprehensive Cancer Center, Chicago, IL 60637, USA
| | - Wei Zhang
- Department of Pediatrics, The University of Illinois at Chicago, Chicago, IL 60607, USA
| | - M Eileen Dolan
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
- The University of Chicago Comprehensive Cancer Center, Chicago, IL 60637, USA
| |
Collapse
|
33
|
Harmonization and semantic annotation of data dictionaries from the Pharmacogenomics Research Network: a case study. J Biomed Inform 2012. [PMID: 23201637 DOI: 10.1016/j.jbi.2012.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The Pharmacogenomics Research Network (PGRN) is a collaborative partnership of research groups funded by NIH to discover and understand how genome contributes to an individual's response to medication. Since traditional biomedical research studies and clinical trials are often conducted independently, common and standardized representations for data are seldom used. This leads to heterogeneity in data representation, which hinders data reuse, data integration and meta-analyses. This study demonstrates harmonization and semantic annotation work for pharmacogenomics data dictionaries collected from PGRN research groups. A semi-automated system was developed to support the harmonization/annotation process, which includes four individual steps, (1) pre-processing PGRN variables; (2) decomposing and normalizing variable descriptions; (3) semantically annotating words and phrases using controlled terminologies; (4) grouping PGRN variables into categories based on the annotation results and semantic types, for total 1514 PGRN variables. Our results demonstrate that there is a significant amount of variability in how pharmacogenomics data is represented and that additional standardization efforts are needed. This represents a critical first step toward identifying and creating data standards for pharmacogenomics studies.
Collapse
|
34
|
Hansson M. Where should we draw the line between quality of care and other ethical concerns related to medical registries and biobanks? THEORETICAL MEDICINE AND BIOETHICS 2012; 33:313-323. [PMID: 22810581 DOI: 10.1007/s11017-012-9229-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Together with large biobanks of human samples, medical registries with aggregated data from many clinical centers are vital parts of an infrastructure for maintaining high standards of quality with regard to medical diagnosis and treatment. The rapid development in personalized medicine and pharmaco-genomics only underscores the future need for these infrastructures. However, registries and biobanks have been criticized as constituting great risks to individual privacy. In this article, I suggest that quality with regard to diagnosis and treatment is an inherent, morally normative requirement of health care, and argue that quality concerns in this sense may be balanced with privacy concerns.
Collapse
Affiliation(s)
- Mats Hansson
- Department of Public Health and Caring Sciences, Centre for Research Ethics & Bioethics, Uppsala University, P.O. Box 564, 751 22, Uppsala, Sweden.
| |
Collapse
|
35
|
Arnedos M, André F, Farace F, Lacroix L, Besse B, Robert C, Soria JC, Eggermont AMM. The challenge to bring personalized cancer medicine from clinical trials into routine clinical practice: the case of the Institut Gustave Roussy. Mol Oncol 2012; 6:204-10. [PMID: 22483534 DOI: 10.1016/j.molonc.2012.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 02/27/2012] [Accepted: 02/27/2012] [Indexed: 11/18/2022] Open
Abstract
Research with high throughput technologies has propitiated the segmentation of different types of tumors into very small subgroups characterized by the presence of very rare molecular alterations. The identification of these subgroups and the apparition of new agents targeting these infrequent alterations are already affecting the way in which clinical trials are being conducted with an increased need to identify those patients harboring specific molecular alterations. In this review we describe some of the currently ongoing and future studies at the Institut Gustave Roussy that aim for the identification of potential therapeutic targets for cancer patients with the incorporation of high throughput technologies into daily practice including aCGH, next generation sequencing and the creation of a software that allows for target identification specific for each tumor. The initial intention is to enrich clinical trials with cancer patients carrying certain molecular alterations in order to increase the possibility of demonstrating benefit from a targeted agent. Mid and long term aims are to facilitate and speed up the process of drug development as well as to implement the concept of personalized medicine.
Collapse
Affiliation(s)
- Monica Arnedos
- Department of Medical Oncology, Institut de Cancerologie Gustave Roussy, 94805 Villejuif, France
| | | | | | | | | | | | | | | |
Collapse
|
36
|
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
|
37
|
Miki I, Nakamura T, Kuwahara A, Yamamori M, Nishiguchi K, Tamura T, Okuno T, Omatsu H, Mizuno S, Hirai M, Azuma T, Sakaeda T. THRB genetic polymorphisms can predict severe myelotoxicity after definitive chemoradiotherapy in patients with esophageal squamous cell carcinoma. Int J Med Sci 2012; 9:748-56. [PMID: 23136537 PMCID: PMC3491433 DOI: 10.7150/ijms.5081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 10/15/2012] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Chemotherapy-related toxicities are difficult to predict before treatment. In this study, we investigated whether thyroid hormone receptor beta (THRB) genetic polymorphisms can serve as a potential biomarker in patients with esophageal squamous cell carcinoma (ESCC). METHODS Forty-nine Japanese patients with ESCC who received a definitive chemoradiotherapy (CRT) with 5-fluorouracil and cisplatin in conjunction with concurrent irradiation were retrospectively analyzed. Severe acute toxicities, including leukopenia, stomatitis, and cheilitis, were evaluated according to 6 single nucleotide polymorphisms (SNPs) in the gene; the intronic SNPs of rs7635707 G/T, rs6787255 A/C, rs9812034 G/T, and rs9310738 C/T and the SNPs in the 3'-untranslated region (3'-UTR) of rs844107 C/T and rs1349265 G/A. RESULTS Distribution of the 4 intronic SNPs, but not the 2 SNPs in the 3'-UTR, showed a significant difference between patients with and without severe acute leukopenia. Stomatitis and cheilitis were not associated with any of the 6 analyzed SNPs. Frequency of haplotype of the 4 intronic SNPs reached approximately 97% with the 2 major haplotypes G-A-G-C (73.4%) and T-C-T-T (23.5%). CONCLUSIONS THRB intronic SNPs can provide useful information on CRT-related severe myelotoxicity in patients with ESCC. Future studies will be needed to confirm these findings.
Collapse
Affiliation(s)
- Ikuya Miki
- Department of Gastroenterology, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|