1
|
Varol A, Klauck SM, Dantzer F, Efferth T. Enhancing cisplatin drug sensitivity through PARP3 inhibition: The influence on PDGF and G-coupled signal pathways in cancer. Chem Biol Interact 2024; 398:111094. [PMID: 38830565 DOI: 10.1016/j.cbi.2024.111094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/07/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
Drug resistance poses a significant challenge in cancer treatment despite the clinical efficacy of cisplatin. Identifying and targeting biomarkers open new ways to improve therapeutic outcomes. In this study, comprehensive bioinformatic analyses were employed, including a comparative analysis of multiple datasets, to evaluate overall survival and mutation hotspots in 27 base excision repair (BER) genes of more than 7,500 tumors across 23 cancer types. By using various parameters influencing patient survival, revealing that the overexpression of 15 distinct BER genes, particularly PARP3, NEIL3, and TDG, consistently correlated with poorer survival across multiple factors such as race, gender, and metastasis. Single nucleotide polymorphism (SNP) analyses within protein-coding regions highlighted the potential deleterious effects of mutations on protein structure and function. The investigation of mutation hotspots in BER proteins identified PARP3 due to its high mutation frequency. Moving from bioinformatics to wet lab experiments, cytotoxic experiments demonstrated that the absence of PARP3 by CRISPR/Cas9-mediated knockdown in MDA-MB-231 breast cancer cells increased drug activity towards cisplatin, carboplatin, and doxorubicin. Pathway analyses indicated the impact of PARP3 absence on the platelet-derived growth factor (PDGF) and G-coupled signal pathways on cisplatin exposure. PDGF, a critical regulator of various cellular functions, was downregulated in the absence of PARP3, suggesting a role in cancer progression. Moreover, the influence of PARP3 knockdown on G protein-coupled receptors (GPCRs) affects their function in the presence of cisplatin. In conclusion, the study demonstrated a synthetic lethal interaction between GPCRs, PDGF signaling pathways, and PARP3 gene silencing. PARP3 emerged as a promising target.
Collapse
Affiliation(s)
- Ayşegül Varol
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University-Mainz, 55128, Mainz, Germany
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ) Heidelberg, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Françoise Dantzer
- Poly(ADP-ribosyl)ation and Genome Integrity, Laboratoire d'Excellence Medalis, UMR7242, Centre Nationale de la Recherche Scientifique/Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, 300 bld. S. Brant, CS10413, 67412, Illkirch, France
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University-Mainz, 55128, Mainz, Germany.
| |
Collapse
|
2
|
Yadav S, Chandra A, Kumar A, Mittal B. Association of TERT-CLPTM1L and 8q24 Common Genetic Variants with Gallbladder Cancer Susceptibility and Prognosis in North Indian Population. Biochem Genet 2018; 56:267-282. [PMID: 29450669 DOI: 10.1007/s10528-018-9843-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 01/27/2018] [Indexed: 12/21/2022]
Abstract
Gallbladder carcinoma (GBC) is one of the common malignancy of the biliary tract. Several genome wide and candidate gene studies have reported associations between multiple cancer types and single-nucleotide polymorphisms on 5p15.33 and 8q24.21 loci. However, predisposition potential of these genetic variants has not been assessed in GBC. We performed the present study to assess the potential of five polymorphisms on 5p15.33 and one on 8q24.21 locus in GBC risk and treatment response in patients undergoing chemoradiotherapy. We extracted genomic DNA from peripheral blood and genotyped selected SNPs using TaqMan allelic discrimination assays in 523 GBC cases and 274 controls from the north-Indian population. Statistical tests were performed to assess the association of selected common genetic variants with gallbladder cancer susceptibility and prognosis. Binary logistic regression analysis showed significant association of TERT rs2736100C > A [OR(CI) = 0.690(0.515-0.924), p value = 0.013], CLPTM1L rs401681C > T [OR(CI) = 0.586(0.405-0.847), p value = 0.004], and CASC8 rs6983267G > T [OR(CI) = 1.629(1.215-2.186), p value = 0.001] with GBC risk. Further, using multivariate logistic regression, we observed that haplotype CLPTM1L Crs401681Crs31489 TERT Trs2853676Ars2736100 MIR4457 Grs4635969 [OR(CI) = 7.52 (1.79-31.52), p value = 0.0064] is significantly associated with poor treatment response. In survival analysis, Kaplan-Meier survival curves showed significantly poor survival and COX regression suggested significantly higher hazard ratio in TT genotype carriers of CASC8 rs6983267 [OR(CI) = 4.28(1. 07-17.10), p value = 0.040] as compared to major allele and heterozygous (GG+GT) genotypes in metastatic GBC cases. The study revealed that 5p15.33 and 8q24.21 genetic variants significantly influence GBC risk and treatment response in north-Indian population.
Collapse
Affiliation(s)
- Saurabh Yadav
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, 226014, India
| | - Abhijit Chandra
- Department of Surgical Gastroenterology, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India
| | - Ashok Kumar
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, 226014, India
| | - Balraj Mittal
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
| |
Collapse
|
3
|
Spracklen TF, Vorster AA, Ramma L, Dalvie S, Ramesar RS. Promoter region variation in NFE2L2 influences susceptibility to ototoxicity in patients exposed to high cumulative doses of cisplatin. THE PHARMACOGENOMICS JOURNAL 2016; 17:515-520. [PMID: 27457817 DOI: 10.1038/tpj.2016.52] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/06/2016] [Accepted: 05/18/2016] [Indexed: 01/09/2023]
Abstract
Ototoxicity is a disabling reaction to cisplatin chemotherapy. Much of the inter-individual variability in the development of hearing impairment among cisplatin-receiving patients has not been fully accounted for. In particular, little is known about the pharmacogenomics of cisplatin-induced ototoxicity. This study sought to investigate the role of variation in five candidate genes in a cohort of South African cancer patients. Five variants within the candidate genes were genotyped in 214 patients, of which SLC22A2 rs316019 and NFE2L2 rs6721961 associated with reduced rates of ototoxicity. In the patients who were exposed to cumulative cisplatin doses ⩾200 mg m-2 (n=113), the variant rs6721961 associated with ototoxicity according to three different grading scales of hearing loss (ASHA, P=0.005; Chang, P=0.028; CTCAE, P=0.004). The NFE2L2 promotor variant rs6721961 may therefore be protective against hearing loss in cisplatin-receiving cancer patients.
Collapse
Affiliation(s)
- T F Spracklen
- MRC Human Genetics Research Unit, Division of Human Genetics, Institute for Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - A A Vorster
- MRC Human Genetics Research Unit, Division of Human Genetics, Institute for Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - L Ramma
- Division of Communication Sciences and Disorders, Groote Schuur Hospital, Cape Town, South Africa
| | - S Dalvie
- Department of Radiation Oncology, Groote Schuur Hospital, Cape Town, South Africa
| | - R S Ramesar
- MRC Human Genetics Research Unit, Division of Human Genetics, Institute for Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
4
|
Travis LB, Fossa SD, Sesso HD, Frisina RD, Herrmann DN, Beard CJ, Feldman DR, Pagliaro LC, Miller RC, Vaughn DJ, Einhorn LH, Cox NJ, Dolan ME. Chemotherapy-induced peripheral neurotoxicity and ototoxicity: new paradigms for translational genomics. J Natl Cancer Inst 2014; 106:dju044. [PMID: 24623533 PMCID: PMC4568989 DOI: 10.1093/jnci/dju044] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 01/22/2014] [Accepted: 01/24/2014] [Indexed: 01/07/2023] Open
Abstract
In view of advances in early detection and treatment, the 5-year relative survival rate for all cancer patients combined is now approximately 66%. As a result, there are more than 13.7 million cancer survivors in the United States, with this number increasing by 2% annually. For many patients, improvements in survival have been countered by therapy-associated adverse effects that may seriously impair long-term functional status, workplace productivity, and quality of life. Approximately 20% to 40% of cancer patients given neurotoxic chemotherapy develop chemotherapy-induced peripheral neurotoxicity (CIPN), which represents one of the most common and potentially permanent nonhematologic side effects of chemotherapy. Permanent bilateral hearing loss and/or tinnitus can result from several ototoxic therapies, including cisplatin- or carboplatin-based chemotherapy. CIPN and ototoxicity represent important challenges because of the lack of means for effective prevention, mitigation, or a priori identification of high-risk patients, and few studies have applied modern genomic approaches to understand underlying mechanisms/pathways. Translational genomics, including cell-based models, now offer opportunities to make inroads for the first time to develop preventive and interventional strategies for CIPN, ototoxicity, and other treatment-related complications. This commentary provides current perspective on a successful research strategy, with a focus on cisplatin, developed by an experienced, transdisciplinary group of researchers and clinicians, representing pharmacogenomics, statistical genetics, neurology, hearing science, medical oncology, epidemiology, and cancer survivorship. Principles outlined herein are applicable to the construction of research programs in translational genomics with strong clinical relevance and highlight unprecedented opportunities to understand, prevent, and treat long-term treatment-related morbidities.
Collapse
Affiliation(s)
- Lois B Travis
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL.
| | - Sophie D Fossa
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Howard D Sesso
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Robert D Frisina
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - David N Herrmann
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Clair J Beard
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Darren R Feldman
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Lance C Pagliaro
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Robert C Miller
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - David J Vaughn
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Lawrence H Einhorn
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - Nancy J Cox
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| | - M Eileen Dolan
- Affiliations of authors: Rubin Center for Cancer Survivorship and Department of Radiation Oncology (LBT) and Department of Neurology (DNH), University of Rochester Medical Center, Rochester, NY; Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway (SDF); Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA (CJB); Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL (RDF); Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY (DRF); Department of Genitourinary Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX (LCP); Department of Radiation Oncology, Mayo Clinic, Rochester, MN (RCM); Department of Medicine, University of Pennsylvania, Philadelphia, PA (DJV); Department of Medical Oncology, Indiana University, Indianapolis, IN (LHE); Departments of Human Genetics (NJC) and Medicine (MED), University of Chicago, Chicago, IL
| |
Collapse
|
5
|
EPS8 inhibition increases cisplatin sensitivity in lung cancer cells. PLoS One 2013; 8:e82220. [PMID: 24367505 PMCID: PMC3868552 DOI: 10.1371/journal.pone.0082220] [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: 06/13/2013] [Accepted: 10/24/2013] [Indexed: 12/15/2022] Open
Abstract
Cisplatin, a commonly used chemotherapeutic, is associated with ototoxicity, renal toxicity and neurotoxicity, thus identifying means to increase the therapeutic index of cisplatin may allow for improved outcomes. A SNP (rs4343077) within EPS8, discovered through a genome wide association study of cisplatin-induced cytotoxicity and apoptosis in lymphoblastoid cell lines (LCLs), provided impetus to further study this gene. The purpose of this work was to evaluate the role of EPS8 in cellular susceptibility to cisplatin in cancerous and non-cancerous cells. We used EPS8 RNA interference to determine the effect of decreased EPS8 expression on LCL and A549 lung cancer cell sensitivity to cisplatin. EPS8 knockdown in LCLs resulted in a 7.9% increase in cisplatin-induced survival (P = 1.98 × 10(-7)) and an 8.7% decrease in apoptosis (P = 0.004) compared to control. In contrast, reduced EPS8 expression in lung cancer cells resulted in a 20.6% decrease in cisplatin-induced survival (P = 5.08 × 10(-5)). We then investigated an EPS8 inhibitor, mithramycin A, as a potential agent to increase the therapeutic index of cisplatin. Mithramycin A decreased EPS8 expression in LCLs resulting in decreased cellular sensitivity to cisplatin as evidenced by lower caspase 3/7 activation following cisplatin treatment (42.7% ± 6.8% relative to control P = 0.0002). In 5 non-small-cell lung carcinoma (NSCLC) cell lines, mithramycin A also resulted in decreased EPS8 expression. Adding mithramycin to 4 NSCLC cell lines and a bladder cancer cell line, resulted in increased sensitivity to cisplatin that was significantly more pronounced in tumor cell lines than in LCL lines (p<0.0001). An EGFR mutant NSCLC cell line (H1975) showed no significant change in sensitivity to cisplatin with the addition of mithramycin treatment. Therefore, an inhibitor of EPS8, such as mithramycin A, could improve cisplatin treatment by increasing sensitivity of tumor relative to normal cells.
Collapse
|
6
|
Mu W, Zhang W. Molecular Approaches, Models, and Techniques in Pharmacogenomic Research and Development. Pharmacogenomics 2013. [DOI: 10.1016/b978-0-12-391918-2.00008-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
7
|
Stark AL, Delaney SM, Wheeler HE, Im HK, Dolan ME. Functional consequences of PRPF39 on distant genes and cisplatin sensitivity. Hum Mol Genet 2012; 21:4348-55. [PMID: 22773733 PMCID: PMC3441128 DOI: 10.1093/hmg/dds266] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 06/15/2012] [Accepted: 06/29/2012] [Indexed: 12/17/2022] Open
Abstract
Variation in gene expression has been found to be important in disease susceptibility and pharmacogenomics. Local and distant expression quantitative trait loci (eQTLs) have been identified via genome-wide association study (GWAS); yet the functional analysis of these variants has been challenging. The aim of this study was to unravel the functional consequence of a gene with a local SNP with evidence for local and distant regulatory roles in cellular sensitivity to cisplatin, one of the most widely used chemotherapeutic drugs. To this end, we measured cellular susceptibility to cisplatin in 176 HapMap lymphoblastoid cell lines derived from Yoruba individuals from Ibadan, Nigeria. The 276 cytotoxicity-associated SNPs at the suggestive threshold of P ≤ 0.0001 were significantly enriched for eQTLs. Of these SNPs, we found one intronic SNP, rs17115814, that had a significant relationship with the expression level of its host gene, PRPF39 (P= 0.0007), and a significant correlation with the expression of over 100 distant transcripts (P ≤ 0.0001). Successful knockdown of PRPF39 expression using siRNA resulted in a significant increase in cisplatin resistance. We then measured the expression of 61 downstream targets after PRPF39 knockdown and found 53 gene targets had significant (P ≤ 0.05) expression changes. Included in the list of genes that significantly changed after PRPF39 knockdown were MAP3K4 and TFPD2, two important signaling genes previously shown to be relevant in cisplatin response. Thus, modulation of a local target gene identified through a GWAS was followed by a downstream cascade of gene expression changes resulting in greater resistance to cisplatin.
Collapse
Affiliation(s)
| | | | | | - Hae Kyung Im
- Department of Health Studies, University of Chicago, Chicago, IL 60637USA
| | - M. Eileen Dolan
- Section of Hematology/Oncology
- Committee on Clinical Pharmacology and Pharmacogenomics, Department of Medicine and
| |
Collapse
|
8
|
Molecular signatures in response to Isoliquiritigenin in lymphoblastoid cell lines. Biochem Biophys Res Commun 2012; 427:392-7. [PMID: 22995316 DOI: 10.1016/j.bbrc.2012.09.070] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 09/12/2012] [Indexed: 11/23/2022]
Abstract
Isoliquiritigenin (ISL) has been known to induce cell cycle arrest and apoptosis of various cancer cells. However, genetic factors regulating ISL effects remain unclear. The aim of this study was to identify the molecular signatures involved in ISL-induced cell death of EBV-transformed lymphoblastoid cell lines (LCLs) using microarray analyses. For gene expression and microRNA (miRNA) microarray experiments, each of 12 LCL strains was independently treated with ISL or DMSO as a vehicle control for a day prior to total RNA extraction. ISL treatment inhibited cell proliferation of LCLs in a dose-dependent manner. Microarray analysis showed that ISL-treated LCLs represented gene expression changes in cell cycle and p53 signaling pathway, having a potential as regulators in LCL survival and sensitivity to ISL-induced cytotoxicity. In addition, 36 miRNAs including five miRNAs with unknown functions were differentially expressed in ISL-treated LCLs. The integrative analysis of miRNA and gene expression profiles revealed 12 putative mRNA-miRNA functional pairs. Among them, miR-1207-5p and miR-575 were negatively correlated with p53 pathway- and cell cycle-associated genes, respectively. In conclusion, our study suggests that miRNAs play an important role in ISL-induced cytotoxicity in LCLs by targeting signaling pathways including p53 pathway and cell cycle.
Collapse
|
9
|
Madian AG, Wheeler HE, Jones RB, Dolan ME. Relating human genetic variation to variation in drug responses. Trends Genet 2012; 28:487-95. [PMID: 22840197 DOI: 10.1016/j.tig.2012.06.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Revised: 06/13/2012] [Accepted: 06/22/2012] [Indexed: 02/03/2023]
Abstract
Although sequencing a single human genome was a monumental effort a decade ago, more than 1000 genomes have now been sequenced. The task ahead lies in transforming this information into personalized treatment strategies that are tailored to the unique genetics of each individual. One important aspect of personalized medicine is patient-to-patient variation in drug response. Pharmacogenomics addresses this issue by seeking to identify genetic contributors to human variation in drug efficacy and toxicity. Here, we present a summary of the current status of this field, which has evolved from studies of single candidate genes to comprehensive genome-wide analyses. Additionally, we discuss the major challenges in translating this knowledge into a systems-level understanding of drug physiology, with the ultimate goal of developing more effective personalized clinical treatment strategies.
Collapse
Affiliation(s)
- Ashraf G Madian
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, IL, USA
| | | | | | | |
Collapse
|
10
|
Yin JY, Huang Q, Zhao YC, Zhou HH, Liu ZQ. Meta-analysis on pharmacogenetics of platinum-based chemotherapy in non small cell lung cancer (NSCLC) patients. PLoS One 2012; 7:e38150. [PMID: 22761669 PMCID: PMC3383686 DOI: 10.1371/journal.pone.0038150] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 05/01/2012] [Indexed: 11/19/2022] Open
Abstract
AIM To determine the pharmacogenetics of platinum-based chemotherapy in Non Small Cell Lung Cancer (NSCLC) patients. METHODS Publications were selected from PubMed, Cochrane Library and ISI Web of Knowledge. A meta-analysis was conducted to determine the association between genetic polymorphisms and platinum-based chemotherapy by checking odds ratio (OR) and 95% confidence interval (CI). RESULTS Data were extracted from 24 publications, which included 11 polymorphisms in 8 genes for meta-analysis. MDR1 C3435T (OR = 1.97, 95% CI: 1.11-3.50, P = 0.02), G2677A/T (OR = 2.61, 95% CI: 1.44-4.74, P = 0.002) and GSTP1 A313G (OR = 0.32, 95% CI: 0.17-0.58, P = 0.0002) were significantly correlated with platinum-based chemotherapy in Asian NSCLC patients. CONCLUSION Attention should be paid to MDR1 C3435T, G2677A/T and GSTP1 A313G for personalized chemotherapy treatment for NSCLC patients in Asian population in the future.
Collapse
Affiliation(s)
- Ji-Ye Yin
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Qiong Huang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Education Ministry, Hefei, Anhui, China
| | - Ying-Chun Zhao
- Osteoporosis Research Center, Creighton University, Omaha, Nebraska, United States of America
| | - Hong-Hao Zhou
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
| | - Zhao-Qian Liu
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China
- * E-mail:
| |
Collapse
|
11
|
Wheeler HE, Dolan ME. Lymphoblastoid cell lines in pharmacogenomic discovery and clinical translation. Pharmacogenomics 2012; 13:55-70. [PMID: 22176622 DOI: 10.2217/pgs.11.121] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The ability to predict how an individual patient will respond to a particular treatment is the ambitious goal of personalized medicine. The genetic make up of an individual has been shown to play a role in drug response. For pharmacogenomic studies, human lymphoblastoid cell lines (LCLs) comprise a useful model system for identifying genetic variants associated with pharmacologic phenotypes. The availability of extensive genotype data for many panels of LCLs derived from individuals of diverse ancestry allows for the study of genetic variants contributing to interethnic and interindividual variation in susceptibility to drugs. Many genome-wide association studies for drug-induced phenotypes have been performed in LCLs, often incorporating gene-expression data. LCLs are also being used in follow-up studies to clinical findings to determine how an associated variant functions to affect phenotype. This review describes the most recent pharmacogenomic findings made in LCLs, including the translation of some findings to clinical cohorts.
Collapse
Affiliation(s)
- Heather E Wheeler
- Section of Hematology/Oncology, Department of Medicine, 900 East 57th St, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
12
|
Tzvetkov MV, Behrens G, O'Brien VP, Hohloch K, Brockmöller J, Benöhr P. Pharmacogenetic analyses of cisplatin-induced nephrotoxicity indicate a renoprotective effect of ERCC1 polymorphisms. Pharmacogenomics 2011; 12:1417-27. [PMID: 21902499 DOI: 10.2217/pgs.11.93] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIM We investigated whether genetic polymorphisms may contribute to the interpatient variability of cisplatin-induced nephrotoxicity. PATIENTS & METHODS Polymorphisms in the candidate genes GSTM1, GSTT1, OCT1, OCT2, LARP2, ERCC1, XRCC1 and EPO were analyzed for associations with nephrotoxicity in 79 cancer patients receiving cisplatin-containing chemotherapy. RESULTS Higher cisplatin dose was associated with strongly decreased estimated glomerular filtration rates (eGFR) (r(2) = 0.205). Two highly genetically linked polymorphisms in the ERCC1 gene, 8092C>A and Asn118Asn, were significantly associated with change in eGFR, accounting for an additional 13% of interindividual variability. Homozygous carriers of the 8092A allele in ERCC1 showed no reduction in eGFR, compared with the 11.5% mean eGFR decrease in C allele carriers (p = 0.004). Homozygous carriers of the C allele of Asn118Asn showed no reduction in eGFR, compared with the 12.8% mean eGFR decrease seen in T allele carriers (p = 0.047). Polymorphisms in the other candidate genes were not associated with cisplatin-induced nephrotoxicity. CONCLUSION Genetic polymorphisms in ERCC1 may be valuable predictors of cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Mladen V Tzvetkov
- Department of Clinical Pharmacology, University Medical Center, Georg-August-University, Robert-Koch-Strasse 40, Göttingen, Germany.
| | | | | | | | | | | |
Collapse
|
13
|
Wen Y, Gorsic LK, Wheeler HE, Ziliak DM, Huang RS, Dolan ME. Chemotherapeutic-induced apoptosis: a phenotype for pharmacogenomics studies. Pharmacogenet Genomics 2011; 21:476-88. [PMID: 21642893 PMCID: PMC3134538 DOI: 10.1097/fpc.0b013e3283481967] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIM To determine whether cellular apoptosis is a suitable phenotypic trait for pharmacogenomics studies by evaluating caspase 3/7-mediated activity in lymphoblastoid cell lines after treatment with six chemotherapeutic agents: 5'-deoxyfluorouridine, pemetrexed, cytarabine, paclitaxel, carboplatin, and cisplatin. MATERIALS AND METHODS Using monozygotic twin pair and sibling pair lymphoblastoid cell lines, we identified conditions for measurement of caspase 3/7 activity in lymphoblastoid cell lines. Genome-wide association studies were performed with over 2 million single nucleotide polymorphisms (SNPs) and cisplatin-induced apoptosis in HapMap CEU cell lines (n=77). RESULTS Although treatment with 5'-deoxyfluorouridine and pemetrexed for up to 24 h resulted in low levels of apoptosis or interindividual variation in caspase-dependent cell death; paclitaxel, cisplatin, carboplatin, and cytarabine treatment for 24 h resulted in 9.4-fold, 9.1-fold, 7.0-fold, and 6.0-fold increases in apoptosis relative to control, respectively. There was a weak correlation between caspase activity and cytotoxicity (r(2)=0.03-0.29) demonstrating that cytotoxicity and apoptosis are two distinct phenotypes that may produce independent genetic associations. Estimated heritability (h(2)) for apoptosis was 0.57 and 0.29 for cytarabine (5 and 40 μmol/l, respectively), 0.22 for paclitaxel (12.5 nmol/l), and 0.34 for cisplatin (5 μmol/l). In the genome-wide association study using the HapMap CEU panel, we identified a significant enrichment of cisplatin-induced cytotoxicity SNPs within the significant cisplatin-induced apoptosis SNPs and an enrichment of expression quantitative trait loci (eQTL). Among these eQTLs, we identified several eQTLs with known function related to apoptosis and/or cytotoxicity. CONCLUSION Our study identifies apoptosis as a phenotype for pharmacogenomic studies in lymphoblastoid cell lines after treatment with paclitaxel, cisplatin, carboplatin, and cytarabine that may have utility for discovering biomarkers to predict response to certain chemotherapeutics.
Collapse
Affiliation(s)
| | | | - Heather E. Wheeler
- Section of Hematology/Oncology, Department of Medicine, the University of Chicago, Chicago, IL 60637, USA
| | - Dana M. Ziliak
- Section of Hematology/Oncology, Department of Medicine, the University of Chicago, Chicago, IL 60637, USA
| | - R. Stephanie Huang
- Section of Hematology/Oncology, Department of Medicine, the University of Chicago, Chicago, IL 60637, USA
| | - M. Eileen Dolan
- Section of Hematology/Oncology, Department of Medicine, the University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
14
|
Tan XL, Moyer AM, Fridley BL, Schaid DJ, Niu N, Batzler AJ, Jenkins GD, Abo RP, Li L, Cunningham JM, Sun Z, Yang P, Wang L. Genetic variation predicting cisplatin cytotoxicity associated with overall survival in lung cancer patients receiving platinum-based chemotherapy. Clin Cancer Res 2011; 17:5801-11. [PMID: 21775533 DOI: 10.1158/1078-0432.ccr-11-1133] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE Inherited variability in the prognosis of lung cancer patients treated with platinum-based chemotherapy has been widely investigated. However, the overall contribution of genetic variation to platinum response is not well established. To identify novel candidate single nucleotide polymorphisms (SNP)/genes, we carried out a genome-wide association study (GWAS) for cisplatin cytotoxicity by using lymphoblastoid cell lines (LCL), followed by an association study of selected SNPs from the GWAS with overall survival (OS) in lung cancer patients. EXPERIMENTAL DESIGN A GWAS for cisplatin was conducted with 283 ethnically diverse LCLs. A total of 168 top SNPs were genotyped in 222 small cell lung cancer (SCLC) and 961 non-SCLC (NSCLC) patients treated with platinum-based therapy. Association of the SNPs with OS was determined by using the Cox regression model. Selected candidate genes were functionally validated by siRNA knockdown in human lung cancer cells. RESULTS Among 157 successfully genotyped SNPs, 9 and 10 SNPs were top SNPs associated with OS for patients with NSCLC and SCLC, respectively, although they were not significant after adjusting for multiple testing. Fifteen genes, including 7 located within 200 kb up or downstream of the 4 top SNPs and 8 genes for which expression was correlated with 3 SNPs in LCLs were selected for siRNA screening. Knockdown of DAPK3 and METTL6, for which expression levels were correlated with the rs11169748 and rs2440915 SNPs, significantly decreased cisplatin sensitivity in lung cancer cells. CONCLUSIONS This series of clinical and complementary laboratory-based functional studies identified several candidate genes/SNPs that might help predict treatment outcomes for platinum-based therapy of lung cancer.
Collapse
Affiliation(s)
- Xiang-Lin Tan
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Cisplatin ototoxicity affects different individuals in a widely variable manner. These variations are likely to be explained by genetic differences among those affected. It would be highly advantageous to identify genetic variants that predispose to cisplatin ototoxicity in order to minimize the risk to susceptible subgroups. Although this area of research is very important, only a few studies have rigorously examined the genetic basis for cisplatin-induced susceptibility to hearing loss. This article addresses recent progress in clarifying the incidence of cisplatin ototoxicity and the risk factors and controversies regarding the identification of genetic variants associated with cisplatin-induced hearing loss.
Collapse
Affiliation(s)
- Debashree Mukherjea
- Department of Surgery, Division of Otolaryngology, Southern Illinois University, School of Medicine, Springfield, IL, USA.
| | | |
Collapse
|
16
|
Wu X, Ye Y, Rosell R, Amos CI, Stewart DJ, Hildebrandt MAT, Roth JA, Minna JD, Gu J, Lin J, Buch SC, Nukui T, Ramirez Serrano JL, Taron M, Cassidy A, Lu C, Chang JY, Lippman SM, Hong WK, Spitz MR, Romkes M, Yang P. Genome-wide association study of survival in non-small cell lung cancer patients receiving platinum-based chemotherapy. J Natl Cancer Inst 2011; 103:817-25. [PMID: 21483023 PMCID: PMC3096796 DOI: 10.1093/jnci/djr075] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 02/02/2011] [Accepted: 02/15/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Interindividual variation in genetic background may influence the response to chemotherapy and overall survival for patients with advanced-stage non-small cell lung cancer (NSCLC). METHODS To identify genetic variants associated with poor overall survival in these patients, we conducted a genome-wide scan of 307,260 single-nucleotide polymorphisms (SNPs) in 327 advanced-stage NSCLC patients who received platinum-based chemotherapy with or without radiation at the University of Texas MD Anderson Cancer Center (the discovery population). A fast-track replication was performed for 315 patients from the Mayo Clinic followed by a second validation at the University of Pittsburgh in 420 patients enrolled in the Spanish Lung Cancer Group PLATAX clinical trial. A pooled analysis combining the Mayo Clinic and PLATAX populations or all three populations was also used to validate the results. We assessed the association of each SNP with overall survival by multivariable Cox proportional hazard regression analysis. All statistical tests were two-sided. RESULTS SNP rs1878022 in the chemokine-like receptor 1 (CMKLR1) was statistically significantly associated with poor overall survival in the MD Anderson discovery population (hazard ratio [HR] of death = 1.59, 95% confidence interval [CI] = 1.32 to 1.92, P = 1.42 × 10(-6)), in the PLATAX clinical trial (HR of death = 1.23, 95% CI = 1.00 to 1.51, P = .05), in the pooled Mayo Clinic and PLATAX validation (HR of death = 1.22, 95% CI = 1.06 to 1.40, P = .005), and in pooled analysis of all three populations (HR of death = 1.33, 95% CI = 1.19 to 1.48, P = 5.13 × 10(-7)). Carrying a variant genotype of rs10937823 was associated with decreased overall survival (HR of death = 1.82, 95% CI = 1.42 to 2.33, P = 1.73 × 10(-6)) in the pooled MD Anderson and Mayo Clinic populations but not in the PLATAX trial patient population (HR of death = 0.96, 95% CI = 0.69 to 1.35). CONCLUSION These results have the potential to contribute to the future development of personalized chemotherapy treatments for individual NSCLC patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Area Under Curve
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/radiotherapy
- Chemotherapy, Adjuvant
- Female
- Genome-Wide Association Study
- Genotype
- Humans
- Kaplan-Meier Estimate
- Linkage Disequilibrium
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Lung Neoplasms/radiotherapy
- Male
- Middle Aged
- Multivariate Analysis
- Neoplasm Staging
- Organoplatinum Compounds/administration & dosage
- Polymorphism, Single Nucleotide
- Proportional Hazards Models
- ROC Curve
- Radiotherapy, Adjuvant
- Receptors, Chemokine/genetics
- Reproducibility of Results
- Smoking/adverse effects
- Smoking/epidemiology
- Treatment Outcome
Collapse
Affiliation(s)
- Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1340, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
The field of pharmacogenomics is focused on the characterization of genetic factors contributing to the response of patients to pharmacological interventions. Drug response and toxicity are complex traits; therefore the effects are likely influenced by multiple genes. The investigation of the genetic basis of drug response has evolved from a focus on single genes to relevant pathways to the entire genome. Preclinical (cell-based models) and clinical genome-wide association studies (GWAS) in oncology provide an unprecedented opportunity for a comprehensive and unbiased assessment of the heritable factors associated with drug response. The primary challenge with attempting to identify pharmacogenomic markers from clinical studies is that they require a homogeneous population of patients treated with the same dosage regimen and minimal confounding variables. Therefore, the development of cell-based models for pharmacogenomic marker identification has utility for the field since performing these types of studies in humans is difficult and costly. This review intends to provide a current report on the status of genomic studies in oncology, the methods for discovery, and implications for patient care. We present a perspective and summary of the challenges and opportunities in translating heritable genomic discoveries to patients.
Collapse
Affiliation(s)
- Federico Innocenti
- Sections of Hematology/Oncology, Department of Medicine, Committee on Clinical Pharmacology and Pharmacogenomics, Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois, USA
| | - Nancy J Cox
- Genetic Medicine, Department of Medicine, Committee on Clinical Pharmacology and Pharmacogenomics, Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois, USA
| | - M Eileen Dolan
- Sections of Hematology/Oncology, Department of Medicine, Committee on Clinical Pharmacology and Pharmacogenomics, Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
18
|
Stark AL, Zhang W, Mi S, Duan S, O’Donnell PH, Huang RS, Dolan ME. Heritable and non-genetic factors as variables of pharmacologic phenotypes in lymphoblastoid cell lines. THE PHARMACOGENOMICS JOURNAL 2010; 10:505-12. [PMID: 20142840 PMCID: PMC2975793 DOI: 10.1038/tpj.2010.3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 11/22/2009] [Accepted: 12/29/2009] [Indexed: 11/09/2022]
Abstract
Publicly available genetic and expression data on lymphoblastoid cell lines (LCLs) make them a unique resource for understanding the genetic underpinnings of pharmacological outcomes and disease. LCLs have been used for pharmacogenomic discovery and validation of clinical findings associated with drug response. However, variation in cellular growth rate, baseline Epstein-Barr virus (EBV) copy number and ATP levels can all be confounders in such studies. Our objective is to better define confounding variables that affect pharmacological end points in LCLs. To this end, we evaluated the effect of these three variables on drug-induced cytotoxicity in LCLs. The drugs evaluated included daunorubicin, etoposide, carboplatin, cisplatin, cytarabine, pemetrexed, 5'-deoxyfluorouridine, vorinostat, methotrexate, 6-mercaptopurine, and 5-fluorouracil. Baseline ATP or EBV copy number were not significantly correlated with cellular growth rate or drug-induced cytotoxicity. In contrast, cellular growth rate and drug-induced cytotoxicity were significantly, directly related for all drugs except vorinostat. Importantly, cellular growth rate is under appreciable genetic influence (h²=0.30-0.39) with five suggestive linkage regions across the genome. Not surprisingly, a percentage of SNPs that significantly associate with drug-induced cytotoxicity also associate with cellular growth rate (P ≤ 0.0001). Studies using LCLs for pharmacologic outcomes should therefore consider that a portion of the genetic variation explaining drug-induced cytotoxicity is mediated via heritable effects on growth rate.
Collapse
Affiliation(s)
- AL Stark
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - W Zhang
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - S Mi
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - S Duan
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - PH O’Donnell
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - RS Huang
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - ME Dolan
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
19
|
Abstract
We review the evidence suggesting the involvement of Cadherin 13 (CDH13, T-cadherin, H-cadherin) in various cancers. CDH13 is an atypical member of the cadherin family, devoid of a transmembrane domain and anchored to the exterior surface of the plasma membrane via a glycosylphosphatidylinositol anchor. CDH13 is thought to affect cellular behavior largely through its signaling properties. It is often down-regulated in cancerous cells. CDH13 down-regulation has been associated with poorer prognosis in various carcinomas, such as lung, ovarian, cervical and prostate cancer. CDH13 re-expression in most cancer cell lines inhibits cell proliferation and invasiveness, increases susceptibility to apoptosis, and reduces tumor growth in in vivo models. These properties suggest that CDH13 may represent a possible target for therapy in some cancers. At the same time, CDH13 is up-regulated in blood vessels growing through tumors and promotes tumor neovascularization. In contrast to most cancer cell lines, CDH13 overexpression in endothelial cells promotes their proliferation and migration, and has a pro-survival effect. We also discuss molecular mechanisms that may regulate CDH13 expression and underlie its roles in cancer.
Collapse
Affiliation(s)
- Alexandra V Andreeva
- Department of Pharmacology, University of Illinois, College of Medicine, Chicago, IL, USA.
| | | |
Collapse
|
20
|
Travis LB, Beard C, Allan JM, Dahl AA, Feldman DR, Oldenburg J, Daugaard G, Kelly JL, Dolan ME, Hannigan R, Constine LS, Oeffinger KC, Okunieff P, Armstrong G, Wiljer D, Miller RC, Gietema JA, van Leeuwen FE, Williams JP, Nichols CR, Einhorn LH, Fossa SD. Testicular cancer survivorship: research strategies and recommendations. J Natl Cancer Inst 2010; 102:1114-30. [PMID: 20585105 DOI: 10.1093/jnci/djq216] [Citation(s) in RCA: 212] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Testicular cancer represents the most curable solid tumor, with a 10-year survival rate of more than 95%. Given the young average age at diagnosis, it is estimated that effective treatment approaches, in particular, platinum-based chemotherapy, have resulted in an average gain of several decades of life. This success, however, is offset by the emergence of considerable long-term morbidity, including second malignant neoplasms, cardiovascular disease, neurotoxicity, nephrotoxicity, pulmonary toxicity, hypogonadism, decreased fertility, and psychosocial problems. Data on underlying genetic or molecular factors that might identify those patients at highest risk for late sequelae are sparse. Genome-wide association studies and other translational molecular approaches now provide opportunities to identify testicular cancer survivors at greatest risk for therapy-related complications to develop evidence-based long-term follow-up guidelines and interventional strategies. We review research priorities identified during an international workshop devoted to testicular cancer survivors. Recommendations include 1) institution of lifelong follow-up of testicular cancer survivors within a large cohort setting to ascertain risks of emerging toxicities and the evolution of known late sequelae, 2) development of comprehensive risk prediction models that include treatment factors and genetic modifiers of late sequelae, 3) elucidation of the effect(s) of decades-long exposure to low serum levels of platinum, 4) assessment of the overall burden of medical and psychosocial morbidity, and 5) the eventual formulation of evidence-based long-term follow-up guidelines and interventions. Just as testicular cancer once served as the paradigm of a curable malignancy, comprehensive follow-up studies of testicular cancer survivors can pioneer new methodologies in survivorship research for all adult-onset cancer.
Collapse
Affiliation(s)
- Lois B Travis
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
O'Donnell PH, Gamazon E, Zhang W, Stark AL, Kistner-Griffin EO, Huang RS, Dolan ME. Population differences in platinum toxicity as a means to identify novel genetic susceptibility variants. Pharmacogenet Genomics 2010; 20:327-37. [PMID: 20393316 PMCID: PMC2908307 DOI: 10.1097/fpc.0b013e3283396c4e] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Clinical studies show that Asians (ASN) are more susceptible to toxicities associated with platinum-containing regimens. We hypothesized that studying ASN as an 'enriched phenotype' population could enable the discovery of novel genetic determinants of platinum susceptibility. METHODS Using well-genotyped lymphoblastoid cell lines from the HapMap, we determined cisplatin and carboplatin cytotoxicity phenotypes (IC50s) for ASN, Caucasians (CEU), and Africans (YRI). IC50s were used in genome-wide association studies. RESULTS ASN were most sensitive to platinums, corroborating clinical findings. ASN genome-wide association studies produced 479 single-nucleotide polymorphisms (SNPs) associating with cisplatin susceptibility and 199 with carboplatin susceptibility (P<10). Considering only the most significant variants (P<9.99x10), backwards elimination was then used to identify reduced-model SNPs, which robustly described the drug phenotypes within ASN. These SNPs comprised highly descriptive genetic signatures of susceptibility, with 12 SNPs explaining more than 95% of the susceptibility phenotype variation for cisplatin, and eight SNPs approximately 75% for carboplatin. To determine the possible function of these variants in ASN, the SNPs were tested for association with differential expression of target genes. SNPs were highly associated with the expression of multiple target genes, and notably, the histone H3 family was implicated for both drugs, suggesting a platinum-class mechanism. Histone H3 has repeatedly been described as regulating the formation of platinum-DNA adducts, but this is the first evidence that specific genetic variants might mediate these interactions in a pharmacogenetic manner. Finally, to determine whether any ASN-identified SNPs might also be important in other human populations, we interrogated all 479/199 SNPs for association with platinum susceptibility in an independent combined CEU/YRI population. Three unique SNPs for cisplatin and 10 for carboplatin replicated in CEU/YRI. CONCLUSION Enriched 'platinum susceptible' populations can be used to discover novel genetic determinants governing interindividual platinum chemotherapy susceptibility.
Collapse
Affiliation(s)
- Peter H. O'Donnell
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois
| | - Eric Gamazon
- Department of Human Genetics, The University of Chicago, Chicago, Illinois
| | - Wei Zhang
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Amy L. Stark
- Department of Human Genetics, The University of Chicago, Chicago, Illinois
| | | | - R. Stephanie Huang
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois
| | - M. Eileen Dolan
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
- Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois
| |
Collapse
|
22
|
Hildebrandt MAT, Gu J, Wu X. Pharmacogenomics of platinum-based chemotherapy in NSCLC. Expert Opin Drug Metab Toxicol 2010; 5:745-55. [PMID: 19442035 DOI: 10.1517/17425250902973711] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
NSCLC is the leading cause of cancer-related death in the US. Patients with NSCLC are mostly treated with platinum-based chemotherapy, often in combination with radiation therapy. However, the development of chemo-resistance is a major hurdle limiting treatment success. In this review, we summarize the current understanding of the genetic factors modulating chemoresistance to platinum chemotherapeutics and their association with clinical outcomes for NSCLC patients. We focus on candidate pathways responsible for drug influx and efflux, metabolism and detoxification, DNA damage repair, and other downstream cellular processes that modulate the effect of platinum-based therapy. We also discuss the application of pathway-based polygenic and genome-wide approaches in identifying genetic factors involved in NSCLC clinical outcomes. Overall, current studies have shown that the effects of each individual polymorphism on clinical outcomes are modest suggesting that a more comprehensive approach that incorporates polygenetic, phenotypic, epidemiologic and clinical variables will be necessary to predict prognosis for NSCLC patients receiving platinum-based chemotherapeutics.
Collapse
Affiliation(s)
- Michelle A T Hildebrandt
- University of Texas M. D. Anderson Cancer Center, Department of Epidemiology, Houston, TX 77030, USA
| | | | | |
Collapse
|
23
|
Morag A, Kirchheiner J, Rehavi M, Gurwitz D. Human lymphoblastoid cell line panels: novel tools for assessing shared drug pathways. Pharmacogenomics 2010; 11:327-40. [DOI: 10.2217/pgs.10.27] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: While powerful in silico tools are emerging for predicting drug targets and pathways, general in vitro tools for assessing such predictions are lacking. We present a novel in vitro method for distinguishing shared versus distinct drug pathways based on comparative cell growth inhibition profiles across a small panel of human lymphoblastoid cell lines (LCLs) from individual donors. Materials & methods: LCLs from unrelated healthy donors were examined in parallel for growth inhibition profiles of various drugs, including antidepressants (paroxetine, fluoxetine, fluvoxamine, citalopram, amitriptyline and imipramine); anticancer drugs (5-fluorouracil, 6-mercaptopurine, azathioprine, methotrexate and resveratrol); steroid drugs (dexamethasone, beclomethasone and prednisolone); and antipsychotic drugs (haloperidol and clozapine). Cell growth was assessed by the colorimetric 2,3-bis(2-methoxy-4-nitro-5-sulfophenly)-5-[(phenylamino) carbonyl]-2H-tetrazolium hydroxide method following 72 h of drug exposure. Results: LCLs from unrelated individuals exhibited a wide range of sensitivities to growth inhibition by a given drug, which were independent of basal cell replication rates. Yet, each individual cell line demonstrated a consistent sensitivity to multiple drugs from the same family. High goodness-of-fit values (R2 > 0.6) were consistently observed for plots comparing the growth-inhibition profiles for paired drugs sharing a similar pathway, for example antidepressants, steroid drugs, antipsychotics, or 6-mercaptopurine compared with azathioprine, but not for drugs with different pathways. The method’s utility is demonstrated by the observation that chlorpheniramine, an antihistamine drug long suspected to also possess antidepressant-like properties, exhibits a growth-inhibition profile very similar to antidepressants. Conclusion: Comparing the growth-inhibition profiles of drugs (or compounds) of interest with the profiles of drugs with known pathways may assist in drug pathway classification. The method is useful for in vitro assessment of in silico-generated drug pathway predictions and for distinguishing shared versus distinct pathways for compounds of interest. Comparative transcriptomics analysis of human lymphoblastoid cell lines exhibiting ‘edge’ sensitivities can subsequently be utilized in the search for drug response biomarkers for personalized pharmacotherapy. The limitations and advantages of the method are discussed.
Collapse
Affiliation(s)
- Ayelet Morag
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Julia Kirchheiner
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, University of Ulm, Ulm, Germany
| | - Moshe Rehavi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - David Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
24
|
Garge N, Pan H, Rowland MD, Cargile BJ, Zhang X, Cooley PC, Page GP, Bunger MK. Identification of quantitative trait loci underlying proteome variation in human lymphoblastoid cells. Mol Cell Proteomics 2010; 9:1383-99. [PMID: 20179311 DOI: 10.1074/mcp.m900378-mcp200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Population-based variability in protein expression patterns, especially in humans, is often observed but poorly understood. Moreover, very little is known about how interindividual genetic variation contributes to protein expression patterns. To begin to address this, we describe elements of technical and biological variations contributing to expression of 544 proteins in a population of 24 individual human lymphoblastoid cell lines that have been extensively genotyped as part of the International HapMap Project. We determined that expression levels of 10% of the proteins were tightly correlated to cell doubling rates. Using the publicly available genotypes for these lymphoblastoid cell lines, we applied a genetic association approach to identify quantitative trait loci associated with protein expression variation. Results identified 24 protein forms corresponding to 15 proteins for which genetic elements were responsible for >50% of the expression variation. The genetic variation associated with protein expression levels were located in cis with the gene coding for the transcript of the protein for 19 of these protein forms. Four of the genetic elements identified were coding non-synonymous single nucleotide polymorphisms that resulted in migration pattern changes in the two-dimensional gel. This is the first description of large scale proteomics analysis demonstrating the direct relationship between genome and proteome variations in human cells.
Collapse
Affiliation(s)
- Nikhil Garge
- Biomarkers and Systems Biology Center, Research Triangle Institute, Research Triangle Park, North Carolina 27709-2194, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Statins, or 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) inhibitors, are widely prescribed to lower plasma cholesterol levels and reduce cardiovascular disease risk. Despite the well-documented efficacy of statins, there is large interindividual variation in response. Using a panel of immortalized lymphocyte cell lines incubated with simvastatin, we recently found that the magnitude of expression of an alternatively spliced HMGCR transcript lacking exon 13 was inversely correlated with in vivo reductions of total cholesterol, low-density lipoprotein cholesterol, apoB, and triglycerides after statin treatment of the individuals from whom the cells were derived. This review will discuss the potential significance of alternative splicing as a mechanism contributing to variation in statin efficacy as well as the use of immortalized lymphocyte cell lines for identifying pharmacogenetically relevant polymorphisms and molecular mechanisms.
Collapse
|
26
|
Wang L. Pharmacogenomics: a systems approach. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2010; 2:3-22. [PMID: 20836007 PMCID: PMC3894835 DOI: 10.1002/wsbm.42] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pharmacogenetics and pharmacogenomics involve the study of the role of inheritance in individual variation in drug response, a phenotype that varies from potentially life-threatening adverse drug reactions to equally serious lack of therapeutic efficacy. Pharmacogenetics-pharmacogenomics represents a major component of the movement to 'individualized medicine'. Pharmacogenetic studies originally focused on monogenic traits, often involving genetic variation in drug metabolism. However, contemporary studies increasingly involve entire 'pathways' that include both pharmacokinetics (PKs)--factors that influence the concentration of a drug reaching its target(s)--and pharmacodynamics (PDs), factors associated with the drug target(s), as well as genome-wide approaches. The convergence of advances in pharmacogenetics with rapid developments in human genomics has resulted in the evolution of pharmacogenetics into pharmacogenomics. At the same time, studies of drug response are expanding beyond genomics to encompass pharmacotranscriptomics and pharmacometabolomics to become a systems-based discipline. This discipline is also increasingly moving across the 'translational interface' into the clinic and is being incorporated into the drug development process and governmental regulation of that process. The article will provide an overview of the development of pharmacogenetics-pharmacogenomics, the scientific advances that have contributed to the continuing evolution of this discipline, the incorporation of transcriptomic and metabolomic data into attempts to understand and predict variation in drug response phenotypes as well as challenges associated with the 'translation' of this important aspect of biomedical science into the clinic.
Collapse
Affiliation(s)
- Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
27
|
Welsh M, Mangravite L, Medina MW, Tantisira K, Zhang W, Huang RS, McLeod H, Dolan ME. Pharmacogenomic discovery using cell-based models. Pharmacol Rev 2009; 61:413-29. [PMID: 20038569 PMCID: PMC2802425 DOI: 10.1124/pr.109.001461] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Quantitative variation in response to drugs in human populations is multifactorial; genetic factors probably contribute to a significant extent. Identification of the genetic contribution to drug response typically comes from clinical observations and use of classic genetic tools. These clinical studies are limited by our inability to control environmental factors in vivo and the difficulty of manipulating the in vivo system to evaluate biological changes. Recent progress in dissecting genetic contribution to natural variation in drug response through the use of cell lines has been made and is the focus of this review. A general overview of current cell-based models used in pharmacogenomic discovery and validation is included. Discussion includes the current approach to translate findings generated from these cell-based models into the clinical arena and the use of cell lines for functional studies. Specific emphasis is given to recent advances emerging from cell line panels, including the International HapMap Project and the NCI60 cell panel. These panels provide a key resource of publicly available genotypic, expression, and phenotypic data while allowing researchers to generate their own data related to drug treatment to identify genetic variation of interest. Interindividual and interpopulation differences can be evaluated because human lymphoblastoid cell lines are available from major world populations of European, African, Chinese, and Japanese ancestry. The primary focus is recent progress in the pharmacogenomic discovery area through ex vivo models.
Collapse
Affiliation(s)
- Marleen Welsh
- Department of Medicine, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Li L, Fridley BL, Kalari K, Jenkins G, Batzler A, Weinshilboum RM, Wang L. Gemcitabine and arabinosylcytosin pharmacogenomics: genome-wide association and drug response biomarkers. PLoS One 2009; 4:e7765. [PMID: 19898621 PMCID: PMC2770319 DOI: 10.1371/journal.pone.0007765] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2009] [Accepted: 10/02/2009] [Indexed: 11/18/2022] Open
Abstract
Cancer patients show large individual variation in their response to chemotherapeutic agents. Gemcitabine (dFdC) and AraC, two cytidine analogues, have shown significant activity against a variety of tumors. We previously used expression data from a lymphoblastoid cell line-based model system to identify genes that might be important for the two drug cytotoxicity. In the present study, we used that same model system to perform a genome-wide association (GWA) study to test the hypothesis that common genetic variation might influence both gene expression and response to the two drugs. Specifically, genome-wide single nucleotide polymorphisms (SNPs) and mRNA expression data were obtained using the Illumina 550K(R) HumanHap550 SNP Chip and Affymetrix U133 Plus 2.0 GeneChip, respectively, for 174 ethnically-defined "Human Variation Panel" lymphoblastoid cell lines. Gemcitabine and AraC cytotoxicity assays were performed to obtain IC(50) values for the cell lines. We then performed GWA studies with SNPs, gene expression and IC(50) of these two drugs. This approach identified SNPs that were associated with gemcitabine or AraC IC(50) values and with the expression regulation for 29 genes or 30 genes, respectively. One SNP in IQGAP2 (rs3797418) was significantly associated with variation in both the expression of multiple genes and gemcitabine and AraC IC(50). A second SNP in TGM3 (rs6082527) was also significantly associated with multiple gene expression and gemcitabine IC50. To confirm the association results, we performed siRNA knock down of selected genes with expression that was associated with rs3797418 and rs6082527 in tumor cell and the knock down altered gemcitabine or AraC sensitivity, confirming our association study results. These results suggest that the application of GWA approaches using cell-based model systems, when combined with complementary functional validation, can provide insights into mechanisms responsible for variation in cytidine analogue response.
Collapse
Affiliation(s)
- Liang Li
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Brooke L. Fridley
- Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Krishna Kalari
- Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Gregory Jenkins
- Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Anthony Batzler
- Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Richard M. Weinshilboum
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Liewei Wang
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
29
|
Fridley BL, Jenkins G, Schaid DJ, Wang L. A Bayesian hierarchical nonlinear model for assessing the association between genetic variation and drug cytotoxicity. Stat Med 2009; 28:2709-22. [PMID: 19572260 PMCID: PMC2755562 DOI: 10.1002/sim.3649] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Non-tumor cell-based model systems have recently gained interest in pharmacogenetic research as a hypothesis generating tool. The hypotheses generated from these model systems can be followed up in functional studies, or tested in individuals taking the same investigational agents. The current cellular phenotypes (e.g. cytotoxicity) of interest in these studies are based on the effects of an individual dosage of a drug on the cell lines, or a summary of results at many dosages of a drug (e.g. dose that inhibits 50 per cent of cell growth, GI 50). A more complete analysis of the impact of genetic variation on all aspects of the dose-response curve may lend additional insight into the pharmacogenomics of a particular drug. This paper illustrates the use of a Bayesian hierarchical nonlinear model for the analysis of pharmacogenomic data with cytotoxicity endpoints. The model is illustrated with cytotoxicity and expression data collected on cell lines from a pharmacogenomic study of the drug gemcitabine. By completing an analysis based on the entire dose-response curve, we were able to detect additional genes that affect not only the GI 50, but also the slope of the curve, which reflects the therapeutic index of the drug. Simulation studies also demonstrate that in comparison with the analyses based on the commonly used summary measure GI 50, investigation of the impact of genetic variation on all aspects of the cytotoxicity dose-response curve is more informative, and more powerful with respect to detecting the effect of gene expression on cytotoxicity.
Collapse
Affiliation(s)
- Brooke L Fridley
- Department of Health Sciences Research, Harwick 766, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|
30
|
Oldenburg J, Fosså SD. Exploration of platinum-induced neuropathy should be based on both objective and subjective examinations. Acta Oncol 2009; 48:804-6. [PMID: 19484592 DOI: 10.1080/02841860902984729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Jan Oldenburg
- Department of Medical Oncology, The Norwegian Radium Hospital, Oslo, Norway
- Buskerud University College, Institute of Health, Drammen, Norway
| | - Sophie D. Fosså
- Department of Clinical Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
- Faculty Division of The Norwegian Radium Hospital, University of Oslo, Oslo, Norway
| |
Collapse
|
31
|
Cavaletti G. Chemotherapy-induced peripheral neurotoxicity: how can we improve knowledge? Lancet Oncol 2009; 10:539-40. [DOI: 10.1016/s1470-2045(09)70105-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
32
|
Kallianpur AR, Hulgan T. Pharmacogenetics of nucleoside reverse-transcriptase inhibitor-associated peripheral neuropathy. Pharmacogenomics 2009; 10:623-37. [PMID: 19374518 PMCID: PMC2706211 DOI: 10.2217/pgs.09.14] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Peripheral neuropathy is an important complication of antiretroviral therapy. Nucleoside reverse-transcriptase inhibitor (NRTI)-associated mitochondrial dysfunction, inflammation and nutritional factors are implicated in its pathogenesis. Pharmacogenetic and genomic studies investigating NRTI neurotoxicity have only recently become possible via the linkage of HIV clinical studies to large DNA repositories. Preliminary case-control studies using these resources suggest that host mitochondrial DNA haplogroup polymorphisms in the hemochromatosis gene and proinflammatory cytokine genes may influence the risk of peripheral neuropathy during antiretroviral therapy. These putative risk factors await confirmation in other HIV-infected populations but they have strong biological plausibility. Work to identify underlying mechanisms for these associations is ongoing. Large-scale studies incorporating clearly defined and validated methods of neuropathy assessment and the use of novel laboratory models of NRTI-associated neuropathy to clarify its pathophysiology are now needed. Such investigations may facilitate the development of more effective strategies to predict, prevent and ameliorate this debilitating treatment toxicity in diverse clinical settings.
Collapse
Affiliation(s)
- Asha R Kallianpur
- Department of Medicine, Vanderbilt University, 2525 West End Avenue, Suite 600, Nashville, TN 37203-31738, USA.
| | | |
Collapse
|
33
|
Bleibel WK, Duan S, Huang RS, Kistner EO, Shukla SJ, Wu X, Badner JA, Dolan ME. Identification of genomic regions contributing to etoposide-induced cytotoxicity. Hum Genet 2009; 125:173-80. [PMID: 19089452 PMCID: PMC2714550 DOI: 10.1007/s00439-008-0607-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Accepted: 12/06/2008] [Indexed: 01/25/2023]
Abstract
Etoposide is routinely used in combination-based chemotherapy for testicular cancer and small-cell lung cancer; however, myelosuppression, therapy-related leukemia and neurotoxicity limit its utility. To determine the genetic contribution to cellular sensitivity to etoposide, we evaluated cell growth inhibition in Centre d' Etude du Polymorphisme Humain lymphoblastoid cell lines from 24 multi-generational pedigrees (321 samples) following treatment with 0.02-2.5 microM etoposide for 72 h. Heritability analysis showed that genetic variation contributes significantly to the cytotoxic phenotypes (h (2) = 0.17-0.25, P = 4.9 x 10(-5)-7.3 x 10(-3)). Whole genome linkage scans uncovered 8 regions with peak LOD scores ranging from 1.57 to 2.55, with the most significant signals being found on chromosome 5 (LOD = 2.55) and chromosome 6 (LOD = 2.52). Linkage-directed association was performed on a subset of HapMap samples within the pedigrees to find 22 SNPs significantly associated with etoposide cytotoxicity at one or more treatment concentrations. UVRAG, a DNA repair gene, SEMA5A, SLC7A6 and PRMT7 are implicated from these unbiased studies. Our findings suggest that susceptibility to etoposide-induced cytotoxicity is heritable and using an integrated genomics approach we identified both genomic regions and SNPs associated with the cytotoxic phenotypes.
Collapse
Affiliation(s)
- Wasim K. Bleibel
- Department of Medicine, University of Chicago, 5841 S. Maryland Ave, Box MC2115, Chicago, IL 60637, USA
| | - Shiwei Duan
- Department of Medicine, University of Chicago, 5841 S. Maryland Ave, Box MC2115, Chicago, IL 60637, USA
| | - R. Stephanie Huang
- Department of Medicine, University of Chicago, 5841 S. Maryland Ave, Box MC2115, Chicago, IL 60637, USA
| | - Emily O. Kistner
- Department of Health Studies, University of Chicago, 5841 S. Maryland Ave, Box MC2115, Chicago, IL, USA
| | - Sunita J. Shukla
- Department of Human Genetics, University of Chicago, 5841 S. Maryland Ave, Box MC2115, Chicago, IL, USA
| | - Xiaolin Wu
- Department of Medicine, University of Chicago, 5841 S. Maryland Ave, Box MC2115, Chicago, IL 60637, USA
| | - Judith A. Badner
- Department of Psychiatry, University of Chicago, 5841 S. Maryland Ave, Box MC2115, Chicago, IL, USA
| | - M. Eileen Dolan
- Department of Medicine, University of Chicago, 5841 S. Maryland Ave, Box MC2115, Chicago, IL 60637, USA
| |
Collapse
|
34
|
Oldenburg J, Fosså SD, Ikdahl T. Genetic variants associated with cisplatin-induced ototoxicity. Pharmacogenomics 2009; 9:1521-30. [PMID: 18855538 DOI: 10.2217/14622416.9.10.1521] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cisplatin induces ototoxicity with a huge interindividual variation, which is at least partly based on genetic differences between the affected individuals. Identification of genetic variants that could predict the severity of ototoxicity is an important step towards a more individualized cisplatin treatment. Nevertheless, so far, only a few studies have assessed this issue. This review will address the prevalence of cisplatin-induced ototoxicity, its pathophysiology, quantification and associations with genetic variants. The recent progress in both phenotyping and genotyping is discussed.
Collapse
Affiliation(s)
- Jan Oldenburg
- Department of Clinical Cancer Research, The Norwegian Radium Hospital, Rikshospitalet, Montebello, 0310 Oslo, Norway.
| | | | | |
Collapse
|
35
|
Shukla SJ, Duan S, Wu X, Badner JA, Kasza K, Dolan ME. Whole-genome approach implicates CD44 in cellular resistance to carboplatin. Hum Genomics 2009; 3:128-42. [PMID: 19164090 PMCID: PMC2683878 DOI: 10.1186/1479-7364-3-2-128] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Accepted: 09/22/2008] [Indexed: 11/10/2022] Open
Abstract
Carboplatin is a chemotherapeutic agent used in the management of many cancers, yet treatment is limited by resistance and toxicities. To achieve a better understanding of the genetic contribution to carboplatin resistance or toxicities, lymphoblastoid cell lines from 34 large Centre d'Etude du Polymorphisme Humain pedigrees were utilised to evaluate interindividual variation in carboplatin cytotoxicity. Significant heritability, ranging from 0.17-0.36 (p = 1 x 10(-7) to 9 x 10(-4)), was found for cell growth inhibition following 72-hour treatment at each carboplatin concentration (10, 20, 40 and 80 microM) and IC(50) (concentration for 50 per cent cell growth inhibition). Linkage analysis revealed 11 regions with logarithm of odds (LOD) scores greater than 1.5. The highest LOD score on chromosome 11 (LOD = 3.36, p = 4.2 x 10(-5)) encompasses 65 genes within the 1 LOD confidence interval for the carboplatin IC 50 . We further analysed the IC(50) phenotype with a linkage-directed association analysis using 71 unrelated HapMap and Perlegen cell lines and identified 18 single nucleotide polymorphisms within eight genes that were significantly associated with the carboplatin IC(50) (p < 3.6 x 10(-5); false discovery rate <5 per cent). Next, we performed linear regression on the baseline expression and carboplatin IC(50) values of the eight associated genes, which identified the most significant correlation between CD44 expression and IC(50) (r(2)= 0.20; p = 6 x 10(-4)). The quantitative real-time polymerase chain reaction further confirmed a statistically significant difference in CD44 expression levels between carboplatin-resistant and -sensitive cell lines (p = 5.9 x 10(-3)). Knockdown of CD44 expression through small interfering RNA resulted in increased cellular sensitivity to carboplatin (p < 0.01). Our whole-genome approach using molecular experiments identified CD44 as being important in conferring cellular resistance to carboplatin.
Collapse
Affiliation(s)
- Sunita J Shukla
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Shiwei Duan
- Department of Medicine, University of Chicago, Chicago IL 60637, USA
| | - Xiaolin Wu
- Department of Medicine, University of Chicago, Chicago IL 60637, USA
| | - Judith A Badner
- Department of Psychiatry, University of Chicago, Chicago IL 60637, USA
| | - Kristen Kasza
- Department of Health Studies, University of Chicago, Chicago IL 60637, USA
| | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago IL 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago IL 60637, USA
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago IL 60637, USA
| |
Collapse
|
36
|
Duan S, Zhang W, Cox NJ, Dolan ME. FstSNP-HapMap3: a database of SNPs with high population differentiation for HapMap3. Bioinformation 2008; 3:139-41. [PMID: 19238253 PMCID: PMC2639690 DOI: 10.6026/97320630003139] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 10/10/2008] [Accepted: 10/15/2008] [Indexed: 11/23/2022] Open
Abstract
The International HapMap Project has recently made available genotypes and frequency data for phase 3 (NCBI build 36,
dbSNPb129) of the HapMap providing an enriched genotype dataset for approximately 1.6 million single nucleotide
polymorphisms (SNPs) from 1,115 individuals with ancestry from parts of Africa, Asia, Europe, North America and Mexico.
In the present study, we aim to facilitate pharmacogenetics studies by providing a database of SNPs with high population
differentiation through a genomewide test on allele frequency variation among 11 HapMap3 samples. Common SNPs with minor
allele frequency greater than 5¢ from each of 11 HapMap3 samples were included in the present analysis. The population
differentiation is measured in terms of fixation index (Fst), and the SNPs with Fst values over 0.5 were defined as highly
differentiated SNPs. Our tests were carried out between all pairs of the 11 HapMap3 samples or among subgroups with the same
continental ancestries. Altogether we carried out 64 genomewide Fst tests and identified 28,215 highly differentiated SNPs
for 49 different combinations of HapMap3 samples in the current database.
Collapse
Affiliation(s)
- Shiwei Duan
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
37
|
Wang L, Weinshilboum RM. Pharmacogenomics: candidate gene identification, functional validation and mechanisms. Hum Mol Genet 2008; 17:R174-9. [PMID: 18852207 PMCID: PMC2574004 DOI: 10.1093/hmg/ddn270] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Accepted: 08/28/2008] [Indexed: 12/19/2022] Open
Abstract
Pharmacogenetics is the study of the role of inheritance in variation in drug response phenotypes. Those phenotypes can range from life-threatening adverse drugs reactions at one end of the spectrum to equally serious lack of therapeutic efficacy at the other. Over the past half century, pharmacogenetics has--like all of medical genetics--evolved from a discipline with a focus on monogenetic traits to become pharmacogenomics, with a genome-wide perspective. This article will briefly review recent examples of the application of genome-wide techniques to clinical pharmacogenomic studies and to pharmacogenomic model systems that vary from cell line-based model systems to yeast gene deletion libraries. Functional validation of candidate genes and the use of genome-wide techniques to gain mechanistic insights will be emphasized for the establishment of biological plausibility and as essential follow-up steps after the identification of candidate genes.
Collapse
Affiliation(s)
| | - Richard M. Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics and Medicine, Mayo Medical School-Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|