Skip to main content
AAPS PharmSci logoLink to AAPS PharmSci
. 2000 Mar 7;2(1):29–41. doi: 10.1208/ps020104

Pharmacogenomics: The promise of personalized medicine

Laviero Mancinelli 1,, Maureen Cronin 1, Wolfgang Sadée 2,
PMCID: PMC2750999  PMID: 11741220

Abstract

Pharmacogenetics and pharmacogenomics deal with the genetic basis underlying variable drug response in individual patients. The traditional pharmacogenetic approach relies on studying sequence variations in candidate genes suspected of affecting drug response. On the other hand, pharmacogenomic studies encompass the sum of all genes, i.e., the genome. Numerous genes may play a role in drug response and toxicity, introducing a daunting level of complexity into the search for candidate genes. The high speed and specificity associated with newly emerging genomic technologies enable the search for relevant genes and their variants to include the entire genome. These new technologies have essentially spawned a new discipline, termed pharmacogenomics, which seeks to identify the variant genes affecting the response to drugs in individual patients. Moreover, pharmacogenomic analysis can identify disease susceptibility genes representing potential new drug targets. All of this will lead to novel approaches in drug discovery, an individualized application of drug therapy, and new insights into disease prevention. Current concepts in drug therapy often attempt treatment of large patient populations as groups, irrespective of the potential for individual, genetically-based differences in drug response. In contrast, pharmacogenomics may help focus effective therapy on smaller patient subpopulations which although demonstrating the same disease phenotype are characterized by distinct genetic profiles. Whether and to what extent this individual, genetics-based approach to medicine results in improved, economically feasible therapy remain to be seen.

To exploit these opportunities in genetic medicine, novel technologies will be needed, legal and ethical questions must be clarified, health care professionals must be educated, and the public must be informed about the implications of genetic testing in drug therapy and disease management.

Key Words: Pharmacogenomics

References

  • 1.Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients. JAMA. 1998;279:1200–1205. doi: 10.1001/jama.279.15.1200. [DOI] [PubMed] [Google Scholar]
  • 2.Sadee W. Finding the right drug for the right patient. Pharm Res. 1998;15:959–963. doi: 10.1023/A:1011949221202. [DOI] [PubMed] [Google Scholar]
  • 3.Maynard Smith J. The Theory of Evolution. Baltimore, Maryland: Penguin Books; 1962. [Google Scholar]
  • 4.Garrod AE. The incidence of alcaptonuria: a study in chemical individuality. Lancet. 1902;ii:1616–1620. doi: 10.1016/S0140-6736(01)41972-6. [DOI] [Google Scholar]
  • 5.Snyder LH. Studies in human inheritance. Ohio J Sci. 1932;32:436–468. [PubMed] [Google Scholar]
  • 6.Ford EB. Genetic Polymorphism. London: Faber & Faber; 1965. [Google Scholar]
  • 7.Nebert DW. Pharmacogenetics: 65 candles on the cake. Pharmacogenetics. 1997;7:435–440. doi: 10.1097/00008571-199712000-00001. [DOI] [Google Scholar]
  • 8.Kalow W. Pharmacogenetics: heredity and the response to drugs. Philadelphia: W. B. Saunders; 1962. [Google Scholar]
  • 9.Carsen PE, Flanagan CL, Iokes CE, Alving AS. Enzymatic deficiency in primaquine-sensitive erythrocytes. Science. 1956;124:484–485. doi: 10.1126/science.124.3220.484-a. [DOI] [PubMed] [Google Scholar]
  • 10.Hughes HB, Biehl JP, Jones AP, Schmidt LH. Metabolism of isoniazid in man as related to the occurrence of peripheral neuritis. Am Rev Tuberculosis. 1954;70:266–273. doi: 10.1164/art.1954.70.2.266. [DOI] [PubMed] [Google Scholar]
  • 11.Evans DAP, Manley KA, McKusick VA. Genetic control of isoniazid metabolism in man. Br Med J. 1960;2:485–490. doi: 10.1136/bmj.2.5197.485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Vatsis K, Martell KJ, Weber WW. Proc Natl Acad Sci U S A. 1991;88:6333–6333. doi: 10.1073/pnas.88.14.6333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Motulsky AG. Drug reactions, enzymes and biochemical genetics. JAMA. 1957;165:835–837. doi: 10.1001/jama.1957.72980250010016. [DOI] [PubMed] [Google Scholar]
  • 14.Eichelbaum M, Steincke B, Dengler JJ. Defective N-oxidation of sparteine in man: a new pharmacogenetic defect. Eur J Clin Pharmacol. 1977;16:183–187. doi: 10.1007/BF00562059. [DOI] [PubMed] [Google Scholar]
  • 15.Weinshilboum RM, Sladek SL. Mercaptopurine pharmacogenetics: monogenic inheritance of erythrocyte thiopurine methyltransferase activity. Am J Hum Genet. 1980;32:651–62. [PMC free article] [PubMed] [Google Scholar]
  • 16.Tai H, Krynetski EY, Yates CR, et al. Thiopurine S-methyltransferase deficiency: two nucleotide transitions define the most prevalent mutant allele associated with loss of catalytic activity in Caucasians. Am J Hum Genet. 1996;58:694–702. [PMC free article] [PubMed] [Google Scholar]
  • 17.Goldenberg MM. Transtuzumab, a recombinant DNA derived humanized monoclonal antibody, a novel agent for the treatment of metastatic breast cancer. Clin Therapeut. 1999;21:309–318. doi: 10.1016/S0149-2918(00)88288-0. [DOI] [PubMed] [Google Scholar]
  • 18.Baselge J, Norton L, Albanell J, Kim YM, Mendelsohn J. Recombinant humanized anti-HER2 antibody (HERCEPTIN) enhances the antitumor activity of paclitaxel and doxorubicin against HER2/neu overexpressing human breast cancer xeno-grafts. Cancer Res. 1998;58:2825–2831. [PubMed] [Google Scholar]
  • 19.Touw DJ. Clinical implications of genetic polymorphisms and drug interactions mediated by cytochrome P450 enzymes. Drug Metab Drug Interact. 1997;14:55–82. [PubMed] [Google Scholar]
  • 20.Evans DAP. Clinical and Molecular Pharmacogenetics. Cambridge: Cambridge University Press; 1993. Genetic factors in drug therapy. [Google Scholar]
  • 21.Buchert E, Woosley RL. Clinical implications of variable antiarrythmic drug metabolism. Pharmacogenet. 1992;2:2–11. doi: 10.1097/00008571-199202000-00002. [DOI] [PubMed] [Google Scholar]
  • 22.Dahl AK, Bertilsson L. Genetically variable metabolism of antidepressants and neuroleptic drugs in man. Pharmacogenet. 1993;3:61–70. doi: 10.1097/00008571-199304000-00001. [DOI] [PubMed] [Google Scholar]
  • 23.Tanaka E, Hisawa S. Clinically significant pharmacokinetic drug interactions with psychoactive drugs: antidepressents and antipsychotics and the cytochrome P450 system. J Clin Pharm Ther. 1999;24:7–16. doi: 10.1046/j.1365-2710.1999.00200.x. [DOI] [PubMed] [Google Scholar]
  • 24.Morais S, Wilkinson GR, Blaisdell J, Nakamura K, Meyer UA, Goldstein JA. The major genetic defect responsible for the polymorphism of S-mephenytoin metabolism in humans. J Biol Chem. 1994;269:15419–15422. [PubMed] [Google Scholar]
  • 25.Daly AK. Molecular basis of polymorphic drug metabolism. J Mol Med. 1995;73:39–553. doi: 10.1007/BF00195139. [DOI] [PubMed] [Google Scholar]
  • 26.Gonzales FJ. Pharmacogenetic phenytyping and genotyping. Present status and future potential. Clin Pharmacokinet. 1994;26:59–70. doi: 10.2165/00003088-199426010-00005. [DOI] [PubMed] [Google Scholar]
  • 27.Ball SE, Scatina JA, Sisenwine SF, Fisher GL. The application of in vitro models of drug metabolism and toxicity in drug discovery and drug development. Drug Chem Toxicity. 1995;18:1–28. doi: 10.3109/01480549509017855. [DOI] [PubMed] [Google Scholar]
  • 28.Guengerich F. The Environmental Genome Project: Functional analysis of polymorphisms. Environment Health Perspect. 1998;106:365–368. doi: 10.2307/3434062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kleyn PW, Vesell ES. Genetic variation as a guide to drug development. Science. 1998;281:1820–1821. doi: 10.1126/science.281.5384.1820. [DOI] [PubMed] [Google Scholar]
  • 30.Sadee W. Pharmacogenomics. BMJ. 1999;319:1286–1286. doi: 10.1136/bmj.319.7220.1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science. 1999;286:487–491. doi: 10.1126/science.286.5439.487. [DOI] [PubMed] [Google Scholar]
  • 32.Iyer VR, Eisen MB, Ross DT, et al. The transcriptional program in the response of human fibroblasts to serum. Science. 1999;283:83–87. doi: 10.1126/science.283.5398.83. [DOI] [PubMed] [Google Scholar]
  • 33.Schachter B. Pharming the Genome. Biomednet [serial online]. October 30, 1998;41.
  • 34.Fluri KG, Fitzpatrick G, Chiem N, Harrison DJ. Integrated capillary electrophoresis devices with an efficient postcolumn reactor in planar quartz and glass chips. Anal Chem. 1996;68:4285–4290. doi: 10.1021/ac9604090. [DOI] [PubMed] [Google Scholar]
  • 35.Jacobson SC, Ramsey SC. Integrated microdevice for DNA restriction fragment analysis. Anal Chem. 1996;68:720–723. doi: 10.1021/ac951230c. [DOI] [PubMed] [Google Scholar]
  • 36.Blanchard AP, Friend SH. Cheap DNA arrays-it’s not all smoke and mirrors. Nature Biotech. 1999;17:953–953. doi: 10.1038/13644. [DOI] [PubMed] [Google Scholar]
  • 37.Russo E.Big pharma hedges its bets The Scientist 1999131–1.12349316 [Google Scholar]
  • 38.Poste G. Bio ’98 International Meeting and Exposition 1–9. New York: RAND Corporation; 1999. [Google Scholar]
  • 39.Weber WW. Pharmacogenetics. New York: Oxford University Press; 1997. [Google Scholar]

Articles from AAPS PharmSci are provided here courtesy of American Association of Pharmaceutical Scientists

RESOURCES