1 / 56

Pharmacogenomics: Clinical Application and Effects on Drug Metabolism

Pharmacogenomics: Clinical Application and Effects on Drug Metabolism. Rodney J. Hunter, Pharm.D. Assistant Professor Texas Southern University College of Pharmacy and Health Sciences SNPhA Regional Conference District III, IV, & V February 19 th , 2011. Financial Disclosure.

heath
Download Presentation

Pharmacogenomics: Clinical Application and Effects on Drug Metabolism

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Pharmacogenomics: Clinical Application and Effects on Drug Metabolism Rodney J. Hunter, Pharm.D. Assistant Professor Texas Southern University College of Pharmacy and Health Sciences SNPhA Regional Conference District III, IV, & V February 19th, 2011

  2. Financial Disclosure I have no conflicts of interest in regards to this program

  3. “Why genetic polymorphisms? Why pharmacogenomics and oncology? Why so much excitement in this field?”

  4. Objectives • Describe pharmacogenomics and the clinical relevance of this field of study in relation to oncology • Outline the drug metabolism pathways affected by pharmacogenomic abnormalities in the oncology patient population • Identify the different drug classes and significant adverse drug effects associated with pharmacogenomic changes in the oncology patient population

  5. Human Genome Project • Begun in 1990 with an expected completion date of 2005, it was completed in 2003 due to advances in technology • Set out to map the 20,000-25,000 human genes • 3 billion DNA bases

  6. Introns and Exons http://www.emc.maricopa.edu .

  7. Genetic Polymorphisms • Single Nucleotide Polymorphism (SNPs) • 1/1000 base pairs • Types of SNPs • Insertion-Deletion • Tandem repeats • Frameshift mutation • Defective splicing • Aberrant splice site • Premature stop codon Dipiro JT, Talbert RL, et al. Pharmacotherapy 7th ed. 2008;6:31-45.

  8. Rationale for Pharmacogenomics • Meta-analysis estimated ~2 million ADR/yr in the US • ADR accounting for 100,000 deaths • Cost in 2000 ~$177.4 billion Lazarou, J et al JAMA. 1998;279:1200-05.

  9. Goals of Pharmacogenomics • Prevent and predict adverse drug reactions • Optimize drug therapy • Lead to novel approaches to drug discovery

  10. Goals of Pharmacogenomics Evans WE and Johnson JA. Annu Rev Genomics Hum Genet. 2001;2:9-39.

  11. Other Pharmacogenomic Implications Evans, WE and McLeod HL. N Engl J Med. 2003;6:538-49.

  12. Pharmacogenomics in Oncology • Drug effects • Drug metabolism • Drug Targets • Drug Transporters • Somatic mutations in malignant tissue • Narrow therapeutic index

  13. Metabolism and Genetics WT/WT = homozygous wild-type allele, Wt/V = heterozygous for one wild-type and one variant allele, V/V = homozygous for variant allele, AUC = area under the curve Evans, WE and McLeod HL. N Engl J Med. 2003;6:538-49.

  14. Efficacy and Genetics WT/WT = homozygous wild-type allele, Wt/V = heterozygous for one wild-type and one variant allele, V/V = homozygous for variant allele, AUC = area under the curve Evans, WE and McLeod HL. N Engl J Med. 2003;6:538-49.

  15. Polygenic Drug Response Evans, WE and McLeod HL. N Engl J Med. 2003;6:538-49.

  16. Thiopurine-S-methyltransferase (TPMT) • Mechanism of Action 6-methylmercaptopurine TPMT 6-thioguanine Azathiopurine 6-mercaptopurine Xanthine Oxidase Thiouric Acid

  17. Thiopurine-S-methyltransferase (TPMT) • TPMT deficiency • Autosomal codominant genetic polymorphism • High TPMT activity is most common • TMPT*3A and TMPT*3C • Heterozygous phenotype • Intermediate tolerance Evans WE, Hon YY, et al. J Clin Oncol. 2001;2293-301.

  18. Thiopurine-S-methyltransferase (TPMT) • Patient population • Patient referred due to excessive toxicity secondary to 6-MP or AZA • Twenty-three patients evaluated • Hospitalizations, platelet transfusions, and missed chemotherapy doses • Genetic Testing • TMPT genotype determined by PCR Evans WE, Hon YY, et al. J Clin Oncol. 2001;2293-301.

  19. 90% Thiopurine-S-methyltransferase (TPMT) Evans WE, Hon YY, et al. J Clin Oncol. 2001;2293-301.

  20. Thiopurine-S-methyltransferase (TPMT) • Summary • A dosage reduction of 90% has benefited patients with homozygous TPMT deficiency • Homozygous TPMT deficient patients have a high incidence of hematopoetic toxicity induced by thiopurines • Heterozygous patients can tolerate full doses in most cases

  21. “JP has a homozygous TPMT deficiency, JP’s full dose of 6-MP is 500 mg/m2/week. What dose should JP receive?”

  22. UGT1A1 • Mechanism of Action

  23. UGT1A1 • UGT1A1 • UGT1A1*28 • Extra TA dinucleotide promotor region • Decreased expression of UGT1A1 protein • Increased risk of irinotecan toxicity • Mainly in homozygous patients Cote JH, Kirzin S, et al. Clin Cancer Res. 2007;3269-75.

  24. UGT1A1 • Patient population • 400 patients with high-risk stage III colon cancer • Patients randomized • 5-fluorouracil/Leucovorin (Arm A) or FOLFIRI (Arm B) • Genetic Testing • DNA from 184 patients was evaluated • UGT1A1, ABCB1, and CYP3A5 Cote JH, Kirzin S, et al. Clin Cancer Res. 2007;3269-75.

  25. UGT1A1 • Results Cote JH, Kirzin S, et al. Clin Cancer Res. 2007;3269-75.

  26. UGT1A1 • Summary • Homozygous patients usually require at least one level dosing reduction • Heterozygous patients can tolerate normal doses • Irinotecan screening • FDA-approved genotype test (Invader® Molecular Assay) Cote JH, Kirzin S, et al. Clin Cancer Res. 2007;3269-75.

  27. Dihydropyrimidine dehydrogenase (DPD) Watters JW, McLeod HL. Biochimica et Biophysica Acta 2003 1603; 99–111

  28. Dihydropyrimidine dehydrogenase (DPD) • DPD deficiency • AG to C single nucleotide change on exon 14 • Decreased activity of DPD responsible for 5-FU breakdown • Major Symptoms • Diarrhea, neutropenia, and neurotoxicity

  29. Thymidylate synthase (TS) Watters JW, McLeod HL. Biochimica et Biophysica Acta 2003 1603; 99–111

  30. Thymidylate synthase (TS) • 5’-promotor enhancer region (TSER) gene

  31. Methylenetetrahydrofolate Reductase (MTHFR) • Mechanism of action in relation to fluoropyrimidines and antifolates • Antifolates • Methotrexate and pemetrexed • Fluoropyrimidines • 5-fluorouracil

  32. Methylenetetrahydrofolate Reductase (MTHFR) • C677T and A1298C • Caucasian and Asian Population • Possible increase susceptibility • 5-FU increased activity • Other implications • Further study mandated • Folate status and geographic origin Dezemtje VO, Guchelaar HJ, et al. Annu Rev Clin Cancer Res 2009;15:15-21.

  33. Thymidylate synthase (TS) • Patient population • 76 patients initiated on 5-FU therapy • TS, DPD, and MTHFR analyzed • Patient Genetic Characteristics • TS 2R/2R – 18%; 2R/3R – 50%; 3R/3R - 30% • MTHFR polymorphisms evenly spread • DPD deficiency in 1.6% Capitain O et al. Pharmacogenomics J 2008;6:256-67.

  34. 5-FU, Genetics, and Outcome • Advanced colorectal cancer patients • Receiving 5-FU (n = 76) • Efficacy • TS 3R/3R genotype (log-rank test p = 0.0065) • Toxicity • DPD (p = 0.031) • MTHFR 1298 A > T (p = 0.0018) Capitain O et al. Pharmacogenomics J 2008;6:256-67.

  35. 5-Fluorouracil (5FU) • Summary • Patients with verified homozygous DPD deficiency should be changed to another treatment regimen • Patients with DPD deficiency develop significantly earlier than patients with normal DPD activity • Increased TS activity linked to poor outcomes in patients treated with 5-FU and its derivatives

  36. “Should RS, a patient with homozygous DPD deficiency be treated with 5-fluorouracil?

  37. Tamoxifen Pharmacogenetics Marsh S and McLeod HL. Expert Opin. Pharmacother. 2007;8:119-27.

  38. CYP 2D6 polymorphism • Mechanism of Action Dezemtje VO, Guchelaar HJ, et al. Annu Rev Clin Cancer Res. 2009;15:15-21.

  39. CYP 2D6 polymorphism • Patient enzyme activity identified by probe • Patients separated in to four groups • Poor, intermediate, extensive, or ultra-rapid metabolizers • Higher rates of recurrence in poor metabolizers Dezemtje VO, Guchelaar HJ, et al. Annu Rev Clin Cancer Res. 2009;15:15-21.

  40. CYP 2D6 polymorphism • Summary • Majority of studies conducted failed to account for tumor grade and prognostic factors • Only one of the major studies accounts for the use of other CYP2D6 inhibitors • Recent studies showing some “negative data” published • AmpliChip CYP450 test is an FDA approved test

  41. Food and Drug Administration • Endocrinology and Metabolic Drugs FDA Advisory Committee • October 16th, 2006 • Package insert updated for tamoxifen “Increased risk of breast cancer recurrence in postmenopausal estrogen receptor positive patients who are CYP2D6 poor metabolizers”

  42. Epidermal Growth Factor Receptor (EGFR) • Small Molecule Inhibitors • Gefitinib and Erlotinib • Monoclonal Antibodies • Cetuximab and Panitumumab • EGFR prognostic or predictive

  43. Epidermal Growth Factor Receptor (EGFR) • Tyrosine Kinase Domain Mutations • Cluster around the ATP-binding sites • Exons 18, 19, and 21 • Most common mutations • L858RA (exon 21) • E19del Yong WO et al. Br J Pharmacol. 2006;62:35-46.

  44. Epidermal Growth Factor Receptor (EGFR) • Major focuses of new research • Somatic mutations tyrosine kinase domain • Erlotinib and Gefitinib • Development of skin rash • Transcriptional activity and expression of EGFR Yong WO et al. Br J Pharmacol. 2006;62:35-46.

  45. Epidermal Growth Factor Receptor (EGFR) • Gefitinib • ISEL • Efficacy associated with nationality • Japanese vs. Caucasian • High gene copy number • Erlotinib • BR.21 • Gene status had no significant contributions to activity

  46. Epidermal Growth Factor Receptor (EGFR) • Patient population • Prospective clinical study • NSCLC, Head and neck CA, and Ovarian CA • Erlotinib • Pharmacokinetics, toxicity, and polymorphic changes • ABCG2 levels Rudin CM et al. J Clin Oncol 2008;26:1119-27.

  47. Phase II studies using EGFR TKIs in EGFR mutation (+) patients Heist RS et al. Pharmacogenomics. 2009;10:59-68.

  48. Kristen-rat Sarcoma (K-Ras) Gene • Mechanism of Action Khambata-Ford S et al. J Clin Oncol. 2007;25:3230-7.

  49. K-Ras • K-Ras mutation • Codon 12 or 13 mutations • G-protein coupled receptor signaling • Cell proliferation and survival • Adenocarcinomas in North America • Progression free survival and overall survival implications?

  50. K-Ras Testing and Economic Implications • Annual incidence of 29,762 mCRC cases • Cetuximab Cost $4,032 loading dose, $2,880/weekly • $753 million annually • K-Ras testing • PCR-based = $452/patient • $13 million annually • Theoretical cost savings of $740 million Shankaran V et al. ASCO GI Cancer Symposium. 2009; abstract 298.

More Related