Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Protein therapeutics: new applications for pharmacogenetics

Key Points

  • Recombinant protein therapeutics have uniquely targeted mechanisms that complement or block endogenous factors. Individual variability in a patient's disease state and genetic background can affect drug responses, which suggests a need to improve methods for patient selection and drug targeting.

  • A target-oriented pharmacogenetic approach is appropriate for highly selective protein drugs, as therapeutic activities, adverse responses and drug elimination are generally mediated through target engagement. However, factors upstream or downstream of the drug target can significantly alter each of these processes.

  • Many pharmacogenetic studies of protein therapeutics have focused on candidate genes within the pharmacological pathways of drug targets. Others have investigated candidate genes associated with disease susceptibility, sub-type or severity. Although exploratory studies have yielded many significant associations, few prospective studies have been performed for protein therapeutics.

  • Genotyping assays are widely available and relatively cheap, but acquiring the resources to develop assays for relevant phenotypic endpoints can be a challenge. In many cases, the functional relevance of (statistically significant) associations between candidate genes and drug response is unknown. A scientific basis for understanding pharmacogenetic associations must be developed, to promote the value of the genetic biomarker.

  • Therapeutics can be derived from endogenous proteins that are subject to genetic variation. Such drugs should be developed with an understanding of the major variants present in the treatment population, to minimize the risk of unintended consequences such as immunogenicity.

  • By integrating pharmacogenetic studies into the early development of new protein therapeutics, scientists can coincidently develop qualified assays for candidate genes and evidence for pharmacogenetic effects on the pharmacodynamic biomarkers typically measured in early-phase clinical studies. With these tools and information in hand, project teams will be poised to derive maximal value from later-phase pharmacogenetic investigations.

Abstract

Pharmacogenetic studies have traditionally focused on genes involved in processes that affect the pharmacokinetics of small-molecule drugs, such as drug metabolism. However, attention is shifting to the effects of genetic variations in drug targets and associated pathway components on drug responses. We describe how these variations are important for understanding differences in responses to the growing number of protein therapeutics that are entering clinical practice. Pharmacogenetic studies of these drugs are surveyed, and issues important to the success of such endeavours are discussed. As novel protein therapeutics are introduced, we anticipate that the use of pharmacogenetics will assume a key role in their development and clinical application.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Time elapsed between FDA approval and first peer-reviewed publication of pharmacogenetic association.

Similar content being viewed by others

References

  1. Salavaggione, O. E., Wang, L., Wiepert, M., Yee, V. C. & Weinshilboum, R. M. Thiopurine S-methyltransferase pharmacogenetics: variant allele functional and comparative genomics. Pharmacogenet. Genomics 15, 801–815 (2005).

    CAS  PubMed  Google Scholar 

  2. Pui, C. H., Relling, M. V. & Evans, W. E. Role of pharmacogenomics and pharmacodynamics in the treatment of acute lymphoblastic leukaemia. Best Pract. Res. Clin. Haematol. 15, 741–756 (2002).

    CAS  PubMed  Google Scholar 

  3. Rieder, M. J. et al. Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. N. Engl. J. Med. 352, 2285–2293 (2005).

    CAS  PubMed  Google Scholar 

  4. Sconce, E. A. et al. The impact of CYP2C9 and VKORC1 genetic polymorphism and patient characteristics upon warfarin dose requirements: proposal for a new dosing regimen. Blood 106, 2329–2333 (2005).

    CAS  PubMed  Google Scholar 

  5. Innocenti, F. et al. Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J. Clin. Oncol. 22, 1382–1388 (2004).

    CAS  PubMed  Google Scholar 

  6. Sadee, W. & Dai, Z. Pharmacogenetics/genomics and personalized medicine. Hum. Mol. Genet. 14, R207–R214 (2005).

    CAS  PubMed  Google Scholar 

  7. Vannier, E. et al. Elevated circulating levels of soluble interleukin-1 receptor type II during interleukin-2 immunotherapy. Eur. Cytokine Netw. 10, 37–42 (1999).

    CAS  PubMed  Google Scholar 

  8. Tilg, H. et al. Induction of circulating antagonists to IL-1 and TNF by IL-2 administration and their effects on IL-2 induced cytokine production in vitro. J. Immunol. 152, 3189–3198 (1994).

    CAS  PubMed  Google Scholar 

  9. Tilg, H. et al. Induction of circulating soluble tumour necrosis factor receptor and interleukin 1 receptor antagonist following interleukin 1 α infusion in humans. Cytokine 6, 215–219 (1994).

    CAS  PubMed  Google Scholar 

  10. Tilg, H., Trehu, E., Atkins, M. B., Dinarello, C. A. & Mier, J. W. Interleukin-6 (IL-6) as an anti-inflammatory cytokine: induction of circulating IL-1 receptor antagonist and soluble tumor necrosis factor receptor p55. Blood 83, 113–118 (1994).

    CAS  PubMed  Google Scholar 

  11. Imanaka, K. et al. Enhanced expression of suppressor of cytokine signalling-1 in the liver of chronic hepatitis C: possible involvement in resistance to interferon therapy. J. Viral Hepat. 12, 130–138 (2005).

    CAS  PubMed  Google Scholar 

  12. Moroni, M. et al. Gene copy number for epidermal growth factor receptor (EGFR) and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. Lancet Oncol. 6, 279–286 (2005).

    CAS  PubMed  Google Scholar 

  13. Bogner, M. P. et al. Serum CD25 levels during interleukin-2 therapy: dose dependence and correlations with clinical toxicity and lymphocyte surface sCD25 expression. J. Immunother. 11, 111–118 (1992).

    CAS  PubMed  Google Scholar 

  14. Sugiyama, Y. Kinetics of receptor-mediated endocytosis of polypeptide hormones that governs the overall hormone disposition in the body: analysis of the uptake process of epidermal growth factor by the liver and kidney. Yakugaku Zasshi 111, 709–736 (1991).

    CAS  PubMed  Google Scholar 

  15. Ng, C. M., Stefanich, E., Anand, B. S., Fielder, P. J. & Vaickus, L. Pharmacokinetics/pharmacodynamics of nondepleting anti-CD4 monoclonal antibody (TRX1) in healthy human volunteers. Pharm. Res. 23, 95–103 (2006). A detailed investigation of the role of receptor-mediated endocytosis in clearance of a monoclonal antibody, this study integrates cell-level and compartmental kinetics into a mechanism-based PK/PD elimination model.

    CAS  PubMed  Google Scholar 

  16. Mager, D. E. & Jusko, W. J. General pharmacokinetic model for drugs exhibiting target-mediated drug disposition. J. Pharmacokinet. Pharmacodyn. 28, 507–532 (2001).

    CAS  PubMed  Google Scholar 

  17. Valentino, L. & Pierre, J. JAK/STAT signal transduction: Regulators and implication in hematological malignancies. Biochem. Pharmacol. 71, 713–721 (2006).

    CAS  PubMed  Google Scholar 

  18. Sjogren, M. H. et al. Interferon alfacon-1 and ribavirin versus interferon α-2b and ribavirin in the treatment of chronic hepatitis C. Dig. Dis. Sci. 50, 727–732 (2005).

    CAS  PubMed  Google Scholar 

  19. Krause, C. D. & Pestka, S. Evolution of the Class 2 cytokines and receptors, and discovery of new friends and relatives. Pharmacol. Ther. 106, 299–346 (2005).

    CAS  PubMed  Google Scholar 

  20. Melian, E. B. & Plosker, G. L. Interferon alfacon-1: a review of its pharmacology and therapeutic efficacy in the treatment of chronic hepatitis C. Drugs 61, 1661–1691 (2001).

    CAS  PubMed  Google Scholar 

  21. Barbosa, M. D., Vielmetter, J., Chu, S., Smith, D. D. & Jacinto, J. Clinical link between MHC class II haplotype and interferon-β (IFN-β) immunogenicity. Clin. Immunol. 118, 42–50 (2005).

    PubMed  Google Scholar 

  22. Fattovich, G. et al. A randomized trial of consensus interferon in combination with ribavirin as initial treatment for chronic hepatitis C. J. Hepatol. 39, 843–849 (2003).

    CAS  PubMed  Google Scholar 

  23. Suzuki, H. & Tango, T. A multicenter, randomized, controlled clinical trial of interferon alfacon-1 in comparison with lymphoblastoid interferon-α in patients with high-titer chronic hepatitis C virus infection. Hepatol. Res. 22, 1–12 (2002).

    CAS  PubMed  Google Scholar 

  24. da Silva, L. C. et al. High rate of sustained response to consensus interferon plus ribavirin in chronic hepatitis C patients resistant to α-interferon and ribavirin: a pilot study. J. Gastroenterol. 37, 732–736 (2002).

    PubMed  Google Scholar 

  25. Koren, E., Zuckerman, L. A. & Mire-Sluis, A. R. Immune responses to therapeutic proteins in humans-clinical significance, assessment and prediction. Curr. Pharm. Biotechnol. 3, 349–360 (2002).

    CAS  PubMed  Google Scholar 

  26. Martin, A. M. et al. Predisposition to abacavir hypersensitivity conferred by HLA-B*5701 and a haplotypic Hsp70-Hom variant. Proc. Natl Acad. Sci. USA 101, 4180–5 (2004).

    CAS  PubMed  Google Scholar 

  27. Hynes, N. E. & Lane, H. A. ERBB receptors and cancer: the complexity of targeted inhibitors. Nature Rev. Cancer 5, 341–354 (2005).

    CAS  Google Scholar 

  28. Pao, W. & Miller, V. A. Epidermal growth factor receptor mutations, small-molecule kinase inhibitors, and non-small-cell lung cancer: current knowledge and future directions. J. Clin. Oncol. 23, 2556–2568 (2005).

    CAS  PubMed  Google Scholar 

  29. Giaccone, G. Epidermal growth factor receptor inhibitors in the treatment of non-small-cell lung cancer. J. Clin. Oncol. 23, 3235–3242 (2005).

    CAS  PubMed  Google Scholar 

  30. Saltz, L. B. et al. Phase II trial of cetuximab in patients with refractory colorectal cancer that expresses the epidermal growth factor receptor. J. Clin. Oncol. 22, 1201–1208 (2004).

    CAS  PubMed  Google Scholar 

  31. Cunningham, D. et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N. Engl. J. Med. 351, 337–345 (2004).

    CAS  PubMed  Google Scholar 

  32. Burtness, B., Goldwasser, M. A., Flood, W., Mattar, B. & Forastiere, A. A. Phase III randomized trial of cisplatin plus placebo compared with cisplatin plus cetuximab in metastatic/recurrent head and neck cancer: an Eastern Cooperative Oncology Group study. J. Clin. Oncol. 23, 8646–8654 (2005).

    PubMed  Google Scholar 

  33. Chung, K. Y. et al. Cetuximab shows activity in colorectal cancer patients with tumors that do not express the epidermal growth factor receptor by immunohistochemistry. J. Clin. Oncol. 23, 1803–1810 (2005).

    CAS  PubMed  Google Scholar 

  34. Mass, R. D. et al. Evaluation of clinical outcomes according to HER2 detection by fluorescence in situ hybridization in women with metastatic breast cancer treated with trastuzumab. Clin. Breast Cancer 6, 240–246 (2005).

    PubMed  Google Scholar 

  35. Dai, Z., Barbacioru, C., Huang, Y. & Sadée, W. Prediction of anticancer drug potency from expression of genes involved in growth factor signaling. Pharm. Res. 23, 336–349 (2006).

    CAS  PubMed  Google Scholar 

  36. Agus, D. B. et al. Phase I clinical study of pertuzumab, a novel HER dimerization inhibitor, in patients with advanced cancer. J. Clin. Oncol. 23, 2534–2543 (2005).

    CAS  PubMed  Google Scholar 

  37. Martinez, A. et al. Association of the major histocompatibility complex with response to infliximab therapy in rheumatoid arthritis patients. Arthritis Rheum. 50, 1077–1082 (2004).

    CAS  PubMed  Google Scholar 

  38. Louis, E. et al. Tumour necrosis factor (TNF) gene polymorphism influences TNF-α production in lipopolysaccharide (LPS)-stimulated whole blood cell culture in healthy humans. Clin. Exp. Immunol. 113, 401–406 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. van Heel, D. A. et al. Inflammatory bowel disease is associated with a TNF polymorphism that affects an interaction between the OCT1 and NFκB transcription factors. Hum. Mol. Genet. 11, 1281–1289 (2002).

    CAS  PubMed  Google Scholar 

  40. Shiina, T., Inoko, H. & Kulski, J. K. An update of the HLA genomic region, locus information and disease associations: 2004. Tissue Antigens 64, 631–649 (2004).

    CAS  PubMed  Google Scholar 

  41. Kang, C. P., Lee, K. W., Yoo, D. H., Kang, C. & Bae, S. C. The influence of a polymorphism at position-857 of the tumour necrosis factor α gene on clinical response to etanercept therapy in rheumatoid arthritis. Rheumatology (Oxford) 44, 547–552 (2005).

    CAS  Google Scholar 

  42. Criswell, L. A. et al. The influence of genetic variation in the HLA-DRB1 and LTA-TNF regions on the response to treatment of early rheumatoid arthritis with methotrexate or etanercept. Arthritis Rheum. 50, 2750–2756 (2004).

    CAS  PubMed  Google Scholar 

  43. Schotte, H. et al. Interleukin 10 promoter microsatellite polymorphisms are associated with response to long term treatment with etanercept in patients with rheumatoid arthritis. Ann. Rheum. Dis. 64, 575–581 (2005).

    CAS  PubMed  Google Scholar 

  44. Hlavaty, T. et al. Polymorphisms in apoptosis genes predict response to infliximab therapy in luminal and fistulizing Crohn's disease. Aliment Pharmacol. Ther. 22, 613–626 (2005).

    CAS  PubMed  Google Scholar 

  45. Camp, N. J. et al. Evidence of a pharmacogenomic response to interleukin-l receptor antagonist in rheumatoid arthritis. Genes Immun. 6, 467–471 (2005).

    CAS  PubMed  Google Scholar 

  46. Marin, F. et al. A pharmacogenetic effect of factor XIII valine 34 leucine polymorphism on fibrinolytic therapy for acute myocardial infarction. J. Am. Coll. Cardiol. 45, 25–29 (2005).

    CAS  PubMed  Google Scholar 

  47. Lim, B. C., Ariens, R. A., Carter, A. M., Weisel, J. W. & Grant, P. J. Genetic regulation of fibrin structure and function: complex gene-environment interactions may modulate vascular risk. Lancet 361, 1424–1431 (2003). A comprehensive in vitro study demonstrates the functional effect of a coding polymorphism on clot structure and presents a medical hypothesis to explain complex clinical pharmacogenetic results.

    CAS  PubMed  Google Scholar 

  48. Horstmann, S., Kalb, P., Koziol, J., Gardner, H. & Wagner, S. Profiles of matrix metalloproteinases, their inhibitors, and laminin in stroke patients: influence of different therapies. Stroke 34, 2165–2170 (2003).

    PubMed  Google Scholar 

  49. Montaner, J. et al. Matrix metalloproteinase-9 pretreatment level predicts intracranial hemorrhagic complications after thrombolysis in human stroke. Circulation 107, 598–603 (2003).

    CAS  PubMed  Google Scholar 

  50. Tsuji, K. et al. Tissue plasminogen activator promotes matrix metalloproteinase-9 upregulation after focal cerebral ischemia. Stroke 36, 1954–1959 (2005).

    CAS  PubMed  Google Scholar 

  51. Montaner, J. et al. Safety profile of tissue plasminogen activator treatment among stroke patients carrying a common polymorphism (C-1562T) in the promoter region of the matrix metalloproteinase-9 gene. Stroke 34, 2851–2855 (2003).

    CAS  PubMed  Google Scholar 

  52. Vallbohmer, D. et al. Molecular determinants of cetuximab efficacy. J. Clin. Oncol. 23, 3536–3544 (2005). Tumour expression level of selected candidate genes by laser capture microdissection was used to identify potential predictive biomarkers for cetuximab efficacy in a small cohort study.

    PubMed  Google Scholar 

  53. Chen, L. et al. Hepatic gene expression discriminates responders and nonresponders in treatment of chronic hepatitis C viral infection. Gastroenterology 128, 1437–1444 (2005).

    CAS  PubMed  Google Scholar 

  54. O'Toole, M. et al. Risk factors associated with β-amyloid(1–42) immunotherapy in preimmunization gene expression patterns of blood cells. Arch. Neurol. 62, 1531–1536 (2005).

    PubMed  Google Scholar 

  55. Sturzebecher, S. et al. Expression profiling identifies responder and non-responder phenotypes to interferon-β in multiple sclerosis. Brain 126, 1419–1429 (2003).

    CAS  PubMed  Google Scholar 

  56. Hong, J., Zang, Y. C., Hutton, G., Rivera, V. M. & Zhang, J. Z. Gene expression profiling of relevant biomarkers for treatment evaluation in multiple sclerosis. J. Neuroimmunol. 152, 126–139 (2004).

    CAS  PubMed  Google Scholar 

  57. Weinstock-Guttman, B. et al. Genomic effects of IFN-β in multiple sclerosis patients. J. Immunol. 171, 2694–2702 (2003). This carefully designed study used GEP to track pharmacodynamic changes in a small cohort; mixed effect models were developed from the time-course data for each gene of interest.

    CAS  PubMed  Google Scholar 

  58. Dall'Ozzo, S. et al. Rituximab-dependent cytotoxicity by natural killer cells: influence of FCGR3A polymorphism on the concentration-effect relationship. Cancer Res. 64, 4664–4669 (2004).

    CAS  PubMed  Google Scholar 

  59. Bowles, J. A. & Weiner, G. J. CD16 polymorphisms and NK activation induced by monoclonal antibody-coated target cells. J. Immunol. Methods 304, 88–99 (2005).

    CAS  PubMed  Google Scholar 

  60. Weng, W. K. & Levy, R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J. Clin. Oncol. 21, 3940–3947 (2003).

    CAS  PubMed  Google Scholar 

  61. Cartron, G. et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcγRIIIa gene. Blood 99, 754–758 (2002).

    CAS  PubMed  Google Scholar 

  62. Farag, S. S. et al. FcγRIIIa and Fcγ RIIa polymorphisms do not predict response to rituximab in B-cell chronic lymphocytic leukemia. Blood 103, 1472–1474 (2004).

    CAS  PubMed  Google Scholar 

  63. Treon, S. P. et al. Polymorphisms in FcγRIIIA (CD16) receptor expression are associated with clinical response to rituximab in Waldenstrom's macroglobulinemia. J. Clin. Oncol. 23, 474–481 (2005).

    CAS  PubMed  Google Scholar 

  64. Lin, T. S. et al. FCGR3A and FCGR2A polymorphisms may not correlate with response to alemtuzumab in chronic lymphocytic leukemia. Blood 105, 289–291 (2005).

    CAS  PubMed  Google Scholar 

  65. Anolik, J. H. et al. The relationship of FcγRIIIa genotype to degree of B cell depletion by rituximab in the treatment of systemic lupus erythematosus. Arthritis Rheum. 48, 455–459 (2003).

    CAS  PubMed  Google Scholar 

  66. Miescher, S. et al. A single recombinant anti-RhD IgG prevents RhD immunization: association of RhD-positive red blood cell clearance rate with polymorphisms in the FcγRIIA and FcγIIIA genes. Blood 103, 4028–4035 (2004).

    CAS  PubMed  Google Scholar 

  67. Hatjiharissi, E. et al. Polymorphisms in FcγRIIIA are genetically linked to FcγRIIA and may account for the primary predictive role ascribed to polymorphisms in FcγRIIIA-158 in determining rituximab responses. Am. Soc. Hematol. Annu. Mtg Abs 106, 2433 (2005).

    Google Scholar 

  68. Louis, E. et al. Association between polymorphism in IgG Fc receptor IIIa coding gene and biological response to infliximab in Crohn's disease. Aliment Pharmacol. Ther. 19, 511–519 (2004).

    CAS  PubMed  Google Scholar 

  69. Willot, S. et al. No association between C-reactive protein gene polymorphisms and decrease of C-reactive protein serum concentration after infliximab treatment in Crohn's disease. Pharmacogenet. Genomics 16, 37–42 (2006).

    CAS  PubMed  Google Scholar 

  70. Tutuncu, Z. et al. Fcγ receptor type IIIA polymorphisms influence treatment outcomes in patients with inflammatory arthritis treated with tumor necrosis factor α-blocking agents. Arthritis Rheum. 52, 2693–2696 (2005).

    CAS  PubMed  Google Scholar 

  71. Koch, K. M. et al. PK and PD of recombinant human IL-18 (rhIL-18) administered IV in repeated cycles to patients with solid tumors. J. Clin. Oncol. 23, 174s A2535 (2005).

    Google Scholar 

  72. Kelly, P. J., Stallard, N. & Whittaker, J. C. Statistical design and analysis of pharmacogenetic trials. Stat. Med. 24, 1495–1508 (2005). A comparison of models to estimate the effect of gene-environment interaction on power to detect a pharmacogenetic effect; models for performing sample size calculations are also presented.

    PubMed  Google Scholar 

  73. Brookes, S. T. et al. Subgroup analyses in randomized trials: risks of subgroup-specific analyses; power and sample size for the interaction test. J. Clin. Epidemiol. 57, 229–236 (2004).

    PubMed  Google Scholar 

  74. Luwor, R. B., Lu, Y., Li, X., Mendelsohn, J. & Fan, Z. The antiepidermal growth factor receptor monoclonal antibody cetuximab/C225 reduces hypoxia-inducible factor-1 α, leading to transcriptional inhibition of vascular endothelial growth factor expression. Oncogene 24, 4433–4441 (2005). An in vitro study elucidates the molecular mechanisms of antiangiogenic effects of cetuximab

    CAS  PubMed  Google Scholar 

  75. Emens, L. A. & Davidson, N. E. Trastuzumab in breast cancer. Oncology (Williston Park) 18, 1117–1128; discussion 1131–1132, 1137–1138 (2004).

    Google Scholar 

  76. Franklin, M. C. et al. Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell 5, 317–328 (2004).

    CAS  PubMed  Google Scholar 

  77. Pao, W. et al. EGF receptor gene mutations are common in lung cancers from 'never smokers' and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc. Natl Acad. Sci. USA 101, 13306–13311 (2004).

    CAS  PubMed  Google Scholar 

  78. Matsuyama, N. et al. The dinucleotide microsatellite polymorphism of the IFNAR1 gene promoter correlates with responsiveness of hepatitis C patients to interferon. Hepatol. Res. 25, 221–225 (2003).

    CAS  PubMed  Google Scholar 

  79. Buckland, P. R. et al. A high proportion of chromosome 21 promoter polymorphisms influence transcriptional activity. Gene Expr. 11, 233–239 (2004).

    PubMed  Google Scholar 

  80. Yoshida, K. et al. Promoter polymorphisms of the interferon-α receptor gene and development of Interferon-induced depressive symptoms in patients with chronic hepatitis C: preliminary findings. Neuropsychobiology 52, 55–61 (2005).

    CAS  PubMed  Google Scholar 

  81. Sriram, U. et al. Pharmacogenomic analysis of interferon receptor polymorphisms in multiple sclerosis. Genes Immun. 4, 147–152 (2003).

    CAS  PubMed  Google Scholar 

  82. Leyva, L. et al. IFNAR1 and IFNAR2 polymorphisms confer susceptibility to multiple sclerosis but not to interferon-β treatment response. J. Neuroimmunol. 163, 165–171 (2005).

    CAS  PubMed  Google Scholar 

  83. Dos Santos, C. et al. A common polymorphism of the growth hormone receptor is associated with increased responsiveness to growth hormone. Nature Genet. 36, 720–724 (2004). The normal distribution in rGH responses was shown to be composed of overlapping growth curves of wild-type children and children carrying a coding polymorphism in GHR; two independent cohorts and in vitro mechanistic studies were used to support hypothesis.

    CAS  PubMed  Google Scholar 

  84. Rowinsky, E. K. et al. Safety, pharmacokinetics, and activity of ABX-EGF, a fully human anti-epidermal growth factor receptor monoclonal antibody in patients with metastatic renal cell cancer. J. Clin. Oncol. 22, 3003–3015 (2004).

    CAS  PubMed  Google Scholar 

  85. Padyukov, L. et al. Genetic markers for the efficacy of tumour necrosis factor blocking therapy in rheumatoid arthritis. Ann. Rheum. Dis. 62, 526–529 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Hughes, L. B. et al. Genetic risk factors for infection in patients with early rheumatoid arthritis. Genes Immun. 5, 641–647 (2004). The effect of multiple alleles that potentially increase infection risk was studied in a large clinical trial; the results suggest additive effects of several genotypes could correlate with adverse events.

    CAS  PubMed  Google Scholar 

  87. Tanaka, S., Edberg, J. C., Chatham, W., Fassina, G. & Kimberly, R. P. FcγRIIIb allele-sensitive release of α-defensins: anti-neutrophil cytoplasmic antibody-induced release of chemotaxins. J. Immunol. 171, 6090–6096 (2003).

    CAS  PubMed  Google Scholar 

  88. Mugnier, B. et al. Polymorphism at position-308 of the tumor necrosis factor α gene influences outcome of infliximab therapy in rheumatoid arthritis. Arthritis Rheum. 48, 1849–1852 (2003).

    CAS  PubMed  Google Scholar 

  89. Fonseca, J. E. et al. Polymorphism at position-308 of the tumour necrosis factor α gene and rheumatoid arthritis pharmacogenetics. Ann. Rheum. Dis. 64, 793–794 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Cuchacovich, M. et al. Tumour necrosis factor-α (TNF-α) levels and influence of-308 TNF-α promoter polymorphism on the responsiveness to infliximab in patients with rheumatoid arthritis. Scand. J. Rheumatol. 33, 228–232 (2004).

    CAS  PubMed  Google Scholar 

  91. Taylor, K. D. et al. ANCA pattern and LTA haplotype relationship to clinical responses to anti-TNF antibody treatment in Crohn's disease. Gastroenterology 120, 1347–1355 (2001).

    CAS  PubMed  Google Scholar 

  92. Louis, E. et al. A positive response to infliximab in Crohn disease: association with a higher systemic inflammation before treatment but not with-308 TNF gene polymorphism. Scand J. Gastroenterol. 37, 818–824 (2002).

    CAS  PubMed  Google Scholar 

  93. Urcelay, E. et al. IBD5 polymorphisms in inflammatory bowel disease: association with response to infliximab. World J. Gastroenterol. 11, 1187–1192 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Mascheretti, S. et al. Pharmacogenetic investigation of the TNF/TNF-receptor system in patients with chronic active Crohn's disease treated with infliximab. Pharmacogenomics J. 2, 127–136 (2002). In this study, a sequential exploratory/confirmatory design was employed to identify and then test associations with candidate genes.

    CAS  PubMed  Google Scholar 

  95. Mascheretti, S. et al. Response to infliximab treatment in Crohn's disease is not associated with mutations in the CARD15 (NOD2) gene: an analysis in 534 patients from two multicenter, prospective GCP-level trials. Pharmacogenetics 12, 509–515 (2002).

    CAS  PubMed  Google Scholar 

  96. Pierik, M. et al. Tumour necrosis factor-α receptor 1 and 2 polymorphisms in inflammatory bowel disease and their association with response to infliximab. Aliment Pharmacol. Ther. 20, 303–310 (2004).

    CAS  PubMed  Google Scholar 

  97. Labbadia, G. et al. Association between human leukocyte antigen (HLA) and interferon- induced thyroid diseases in four patients with HCV-related chronic hepatitis. Neuro. Endocrinol. Lett. 26, 109–112 (2005).

    CAS  PubMed  Google Scholar 

  98. Floreani, A. et al. Organ-specific autoimmunity and genetic predisposition in interferon-treated HCV-related chronic hepatitis patients. Ital J. Gastroenterol. Hepatol 30, 71–76 (1998).

    CAS  Google Scholar 

  99. Knapp, S. et al. Interleukin-10 promoter polymorphisms and the outcome of hepatitis C virus infection. Immunogenetics 55, 362–369 (2003).

    CAS  PubMed  Google Scholar 

  100. Suzuki, F. et al. Single nucleotide polymorphism of the MxA gene promoter influences the response to interferon monotherapy in patients with hepatitis C viral infection. J. Viral Hepat. 11, 271–276 (2004).

    CAS  PubMed  Google Scholar 

  101. Knapp, S. et al. Polymorphisms in interferon-induced genes and the outcome of hepatitis C virus infection: roles of MxA, OAS-1 and PKR. Genes Immun. 4, 411–419 (2003).

    CAS  PubMed  Google Scholar 

  102. Konishi, I., Horiike, N., Hiasa, Y., Michitaka, K. & Onji, M. CCR5 promoter polymorphism influences the interferon response of patients with chronic hepatitis C in Japan. Intervirology 47, 114–120 (2004).

    CAS  PubMed  Google Scholar 

  103. Mascheretti, S. et al. Genetic variants in the CCR gene cluster and spontaneous viral elimination in hepatitis C-infected patients. Clin. Exp. Immunol. 136, 328–333 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Schiemann, U. et al. Response to combination therapy with interferon alfa-2a and ribavirin in chronic hepatitis C according to a TNF-α promoter polymorphism. Digestion 68, 1–4 (2003).

    CAS  Google Scholar 

  105. Dai, C. Y. et al. Polymorphisms in the interferon-γ gene at position +874 in patients with chronic hepatitis C treated with high-dose interferon-alpha and ribavirin. Antiviral Res. 67, 93–97 (2005).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors wish to thank our colleagues for helpful discussion and critical review of this manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Cecile Krejsa.

Ethics declarations

Competing interests

C.K. and M.R. are employees of ZymoGenetics, Inc. which is involved in the production and commercialization of therapeutic proteins for the prevention and treatment of human diseases.

Related links

Related links

DATABASES

OMIM

Chronic lymphocytic leukaemia

Crohn's disease

follicular lymphoma

multiple sclerosis

psoriasis

rheumatoid arthritis

stroke

Waldrenstrom's macroglobulinaemia

FURTHER INFORMATION

Draft Guidance for Industry and FDA Staff-Pharmacogenetic Tests and Genetic Tests for Heritable Markers

EMEA Guideline on Pharmacogenetics Briefing Meetings

EMEA Position Paper on Terminology in Pharmacogenetics

US FDA Guidance for Industry Pharmacogenomic Data Submissions

US FDA Pharmacogenomic Data Submission

Glossary

Recombinant protein therapeutics

Include cytokines, growth factors, enzymes, coagulation factors, soluble receptors, monoclonal antibodies and engineered forms of these proteins.

Polymorphism

A genetic variation in the DNA sequence with a measurable frequency of detection above 1%. Any polymorphism in the chromosomal DNA (coding and non-coding) can affect mRNA processing, maturation and translation, as a result of changes in conformation. A subset of polymorphisms alters the translated protein.

Single nucleotide polymorphism (SNP)

A specific location in a DNA sequence at which different people can have a different DNA base. SNPs can occur in coding or non-coding regions of a gene; a coding SNP can be synonymous (the codon encodes the same amino acid), non-synonymous (an amino acid change) or nonsense (a premature stop results).

Microsatellite repeat

A class of repetitive DNA that is made up of repeats that are two to eight nucleotides in length. They can be highly polymorphic and are frequently used as molecular markers in population genetics studies.

Genotype

The set of two alleles carried by an individual at a given polymorphic site.

Haplotype

The composite of several alleles carried by an individual across a region of DNA. Haplotypes can be constructed from multiple polymorphic alleles observed in a single gene, or extended to incorporate polymorphic sites across several genes.

Diplotype

The combination of two haplotypes carried by an individual.

Phenotype

The physical manifestation of genetic traits, or general constitutional manifestation of health or disease in an individual. A patients phenotype can describe disease status, but phenotype can also relate to specific responses such as analytical test results.

Pharmacokinetics

Pharmacokinetics describes the absorption, distribution, metabolism and excretion of the drug, and yields an estimate of drug exposure.

Pharmacodynamics

Drug pharmacodynamics generally uses biomarkers to monitor the biological response to drug–target interaction.

Dominant model

In the dominant model, carriage of a single copy of the allele has the same functional outcome as carriage of two copies (the allele is dominant).

Recessive model

In the recessive model, carriage of a single copy of the allele has the same functional outcome as carriage of zero copies of the variant allele (the allele is recessive).

Additive model

In the additive model, carriage of a single copy of the allele has functional effects intermediate to carriage of zero or two copies (the alleles are additive and gene-dosage effects might apply).

Rights and permissions

Reprints and permissions

About this article

Cite this article

Krejsa, C., Rogge, M. & Sadee, W. Protein therapeutics: new applications for pharmacogenetics. Nat Rev Drug Discov 5, 507–521 (2006). https://doi.org/10.1038/nrd2039

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd2039

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing