1932

Abstract

The immunological mechanisms driving delayed hypersensitivity reactions (HSRs) to drugs mediated by drug-reactive T lymphocytes are exemplified by several key examples and their human leukocyte antigen (HLA) associations: abacavir and , carbamazepine and , allo-purinol and , and both amoxicillin-clavulanate and nevirapine with multiple class I and II alleles. For HLA-restricted drug HSRs, specific class I and/or II HLA alleles are necessary but not sufficient for tissue specificity and the clinical syndrome. Several models have been proposed to explain the immunopathogenesis of severe T cell–mediated drug HSRs, and our increased understanding of the risk factors and mechanisms involved in the development of these reactions will further the development of sensitive and specific strategies for preclinical screening that will lead to safer and more cost-effective drug design.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-050913-022745
2015-01-14
2024-04-26
Loading full text...

Full text loading...

/deliver/fulltext/med/66/1/annurev-med-050913-022745.html?itemId=/content/journals/10.1146/annurev-med-050913-022745&mimeType=html&fmt=ahah

Literature Cited

  1. Gell PGH, Coombs RRA. 1.  1963. The classification of allergic reactions underlying disease. Clinical Aspects of Immunology RRA Coombs, PGH Gell 317–37 London: Blackwell Sci. [Google Scholar]
  2. Pirmohamed M, Aithal GP, Behr E. 2.  et al. 2011. The phenotype standardization project: improving pharmacogenetic studies of serious adverse drug reactions. Clin. Pharmacol. Ther. 89:784–85 [Google Scholar]
  3. Hung SI, Chung WH, Liou LB. 3.  et al. 2005. HLA-B*5801 allele as a genetic marker for severe cutaneous adverse reactions caused by allopurinol. Proc. Natl. Acad. Sci. USA 102:4134–39 [Google Scholar]
  4. Somkrua R, Eickman EE, Saokaew S. 4.  et al. 2011. Association of HLA-B*5801 allele and allopurinol-induced Stevens Johnson syndrome and toxic epidermal necrolysis: a systematic review and meta-analysis. BMC Med. Genet. 12:118 [Google Scholar]
  5. Zhang FR, Liu H, Irwanto A. 5.  et al. 2013. HLA-B*13:01 and the dapsone hypersensitivity syndrome. N. Engl. J. Med. 369:1620–28 [Google Scholar]
  6. Daly AK, Donaldson PT, Bhatnagar P. 6.  et al. 2009. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat. Genet. 41:816–19 [Google Scholar]
  7. Hetherington S, Hughes AR, Mosteller M. 7.  et al. 2002. Genetic variations in HLA-B region and hypersensitivity reactions to abacavir. Lancet 359:1121–22 [Google Scholar]
  8. Mallal S, Nolan D, Witt C. 8.  et al. 2002. Association between presence of HLA-B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir. Lancet 359:727–32 [Google Scholar]
  9. Chung WH, Hung SI, Hong HS. 9.  et al. 2004. Medical genetics: a marker for Stevens-Johnson syndrome. Nature 428:486 [Google Scholar]
  10. Hung SI, Chung WH, Jee SH. 10.  et al. 2006. Genetic susceptibility to carbamazepine-induced cutaneous adverse drug reactions. Pharmacogenet. Genomics 16:297–306 [Google Scholar]
  11. McCormack M, Alfirevic A, Bourgeois S. 11.  et al. 2011. HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans. N. Engl. J. Med. 364:1134–43 [Google Scholar]
  12. Hautekeete ML, Horsmans Y, Van Waeyenberge C. 12.  et al. 1999. HLA association of amoxicillin-clavulanate–induced hepatitis. Gastroenterology 117:1181–86 [Google Scholar]
  13. Donaldson PT, Daly AK, Henderson J. 13.  et al. 2010. Human leucocyte antigen class II genotype in susceptibility and resistance to co-amoxiclav-induced liver injury. J. Hepatol. 53:1049–53 [Google Scholar]
  14. O'Donohue J, Oien KA, Donaldson P. 14.  et al. 2000. Co-amoxiclav jaundice: clinical and histological features and HLA class II association. Gut 47:717–20 [Google Scholar]
  15. Martin AM, Nolan D, James I. 15.  et al. 2005. Predisposition to nevirapine hypersensitivity associated with HLA-DRB1*0101 and abrogated by low CD4 T-cell counts. AIDS 19:97–99 [Google Scholar]
  16. Yuan J, Guo S, Hall D. 16.  et al. 2011. Toxicogenomics of nevirapine-associated cutaneous and hepatic adverse events among populations of African, Asian, and European descent. AIDS 25:1271–80 [Google Scholar]
  17. Spraggs CF, Budde LR, Briley LP. 17.  et al. 2011. HLA-DQA1*02:01 is a major risk factor for lapatinib-induced hepatotoxicity in women with advanced breast cancer. J. Clin. Oncol. 29:667–73 [Google Scholar]
  18. Spraggs CF, Parham LR, Hunt CM, Dollery CT. 18.  2012. Lapatinib-induced liver injury characterized by class II HLA and Gilbert's syndrome genotypes. Clin. Pharmacol. Ther. 91:647–52 [Google Scholar]
  19. Singer JB, Lewitzky S, Leroy E. 19.  et al. 2010. A genome-wide study identifies HLA alleles associated with lumiracoxib-related liver injury. Nat. Genet. 42:711–14 [Google Scholar]
  20. Keisu M, Andersson TB. 20.  2010. Drug-induced liver injury in humans: the case of ximelagatran. Handb. Exp. Pharmacol. 196:407–18 [Google Scholar]
  21. Yvon M, Anglade P, Wal JM. 21.  1990. Identification of the binding sites of benzyl penicilloyl, the allergenic metabolite of penicillin, on the serum albumin molecule. FEBS Lett. 263:237–40 [Google Scholar]
  22. Kelly JW, Dooley DP, Lattuada CP, Smith CE. 22.  1992. A severe, unusual reaction to trimethoprim-sulfamethoxazole in patients infected with human immunodeficiency virus. Clin. Infect. Dis. 14:1034–39 [Google Scholar]
  23. Pichler WJ, Beeler A, Keller M. 23.  et al. 2006. Pharmacological interaction of drugs with immune receptors: the P-I concept. Allergol. Int. 55:17–25 [Google Scholar]
  24. Cutrell AG, Hernandez JE, Fleming JW. 24.  et al. 2004. Updated clinical risk factor analysis of suspected hypersensitivity reactions to abacavir. Ann. Pharmacother. 38:2171–72 [Google Scholar]
  25. Rauch A, Nolan D, Thurnheer C. 25.  et al. 2008. Refining abacavir hypersensitivity diagnoses using a structured clinical assessment and genetic testing in the Swiss HIV Cohort Study. Antivir. Ther. 13:1019–28 [Google Scholar]
  26. Saag M, Balu R, Phillips E. 26.  et al. 2008. High sensitivity of human leukocyte antigen-B*5701 as a marker for immunologically confirmed abacavir hypersensitivity in white and black patients. Clin. Infect. Dis. 46:1111–18 [Google Scholar]
  27. Mallal S, Phillips E, Carosi G. 27.  et al. 2008. HLA-B*5701 screening for hypersensitivity to abacavir. N. Engl. J. Med. 358:568–79 [Google Scholar]
  28. Martin AM, Nolan D, Gaudieri S. 28.  et al. 2004. Predisposition to abacavir hypersensitivity conferred by HLA-B*5701 and a haplotypic Hsp70-Hom variant. Proc. Natl. Acad. Sci. USA 101:4180–85 [Google Scholar]
  29. Rauch A, Nolan D, Martin A. 29.  et al. 2006. Prospective genetic screening decreases the incidence of abacavir hypersensitivity reactions in the Western Australian HIV cohort study. Clin. Infect. Dis. 43:99–102 [Google Scholar]
  30. Chessman D, Kostenko L, Lethborg T. 30.  et al. 2008. Human leukocyte antigen class I-restricted activation of CD8+ T cells provides the immunogenetic basis of a systemic drug hypersensitivity. Immunity 28:822–32 [Google Scholar]
  31. Illing PT, Vivian JP, Dudek NL. 31.  et al. 2012. Immune self-reactivity triggered by drug-modified HLA-peptide repertoire. Nature 486:554–58 [Google Scholar]
  32. Ostrov DA, Grant BJ, Pompeu YA. 32.  et al. 2012. Drug hypersensitivity caused by alteration of the MHC-presented self-peptide repertoire. Proc. Natl. Acad. Sci. USA 109:9959–64 [Google Scholar]
  33. Norcross MA, Luo S, Lu L. 33.  et al. 2012. Abacavir induces loading of novel self-peptides into HLA-B*57:01: an autoimmune model for HLA-associated drug hypersensitivity. AIDS 26:F21–29 [Google Scholar]
  34. Amir AL, D'Orsogna LJ, Roelen DL. 34.  et al. 2010. Allo-HLA reactivity of virus-specific memory T cells is common. Blood 115:3146–57 [Google Scholar]
  35. Amir AL, van der Steen DM, Hagedoorn RS. 35.  et al. 2011. Allo-HLA-reactive T cells inducing graft-versus-host disease are single peptide specific. Blood 118:6733–42 [Google Scholar]
  36. Burrows SR, Khanna R, Burrows JM, Moss DJ. 36.  1994. An alloresponse in humans is dominated by cytotoxic T lymphocytes (CTL) cross-reactive with a single Epstein-Barr virus CTL epitope: implications for graft-versus-host disease. J. Exp. Med. 179:1155–61 [Google Scholar]
  37. Gamadia LE, Remmerswaal EB, Surachno S. 37.  et al. 2004. Cross-reactivity of cytomegalovirus-specific CD8+ T cells to allo–major histocompatibility complex class I molecules. Transplantation 77:1879–85 [Google Scholar]
  38. Koelle DM, Chen HB, McClurkan CM, Petersdorf EW. 38.  2002. Herpes simplex virus type 2–specific CD8 cytotoxic T lymphocyte cross-reactivity against prevalent HLA class I alleles. Blood 99:3844–47 [Google Scholar]
  39. Deckers JGM, Boonstra JG, Van Der Kooij SW. 39.  et al. 1997. Tissue-specific characteristics of cytotoxic graft-infiltrating T cells during renal allograft rejection. Transplantation 64:178–81 [Google Scholar]
  40. D'Orsogna LJ, Roelen DL, Doxiadis II, Claas FH. 40.  2010. Alloreactivity from human viral specific memory T-cells. Transpl. Immunol. 23:149–55 [Google Scholar]
  41. D'Orsogna LJ, Roelen DL, van der Meer-Prins EM. 41.  et al. 2011. Tissue specificity of cross-reactive allogeneic responses by EBV EBNA3A-specific memory T cells. Transplantation 91:494–500 [Google Scholar]
  42. Zhu J, Peng T, Johnston C. 42.  et al. 2013. Immune surveillance by CD8αα+ skin-resident T cells in human herpes virus infection. Nature 497:494–97 [Google Scholar]
  43. Kaniwa N, Saito Y, Aihara M. 43.  et al. 2010. HLA-B*1511 is a risk factor for carbamazepine-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in Japanese patients. Epilepsia 51:2461–65 [Google Scholar]
  44. Sun D, Yu CH, Liu ZS. 44.  et al. 2014. Association of HLA-B*1502 and *1511 allele with antiepileptic drug-induced Stevens-Johnson syndrome in central China. J. Huazhong Univ. Sci. Technol. 34:146–50 [Google Scholar]
  45. Hung SI, Chung WH, Liu ZS. 45.  et al. 2010. Common risk allele in aromatic antiepileptic-drug induced Stevens-Johnson syndrome and toxic epidermal necrolysis in Han Chinese. Pharmacogenomics 11:349–56 [Google Scholar]
  46. Wu Y, Wang XQ, Gan ZJ, Fan M. 46.  2012. HLA-B*1502 allele screening: determination of carbamazepine-induced cutaneous reactions. Epilepsia 53:929–30 [Google Scholar]
  47. Ozeki T, Mushiroda T, Yowang A. 47.  et al. 2011. Genome-wide association study identifies HLA-A*3101 allele as a genetic risk factor for carbamazepine-induced cutaneous adverse drug reactions in Japanese population. Hum. Mol. Genet. 20:1034–41 [Google Scholar]
  48. Grover S, Kukreti R. 48.  2014. HLA alleles and hypersensitivity to carbamazepine: an updated systematic review with meta-analysis. Pharmacogenet. Genomics 24:94–112 [Google Scholar]
  49. Genin E, Chen DP, Hung SI. 49.  et al. 2014. HLA-A*31:01 and different types of carbamazepine-induced severe cutaneous adverse reactions: an international study and meta-analysis. Pharmacogenomics J. 14:281–88 [Google Scholar]
  50. Amstutz U, Ross CJ, Castro-Pastrana LI. 50.  et al. 2013. HLA-A 31:01 and HLA-B 15:02 as genetic markers for carbamazepine hypersensitivity in children. Clin. Pharmacol. Ther. 94:142–49 [Google Scholar]
  51. Niihara H, Kakamu T, Fujita Y. 51.  et al. 2011. HLA-A31 strongly associates with carbamazepine-induced adverse drug reactions but not with carbamazepine-induced lymphocyte proliferation in a Japanese population. J. Dermatol. 39:594–601 [Google Scholar]
  52. Lichtenfels M, Farrell J, Ogese MO. 52.  et al. 2014. HLA restriction of carbamazepine-specific T-cell clones from an HLA-A*31:01-positive hypersensitive patient. Chem. Res. Toxicol. 27:175–77 [Google Scholar]
  53. Ko TM, Chung WH, Wei CY. 53.  et al. 2011. Shared and restricted T-cell receptor use is crucial for carbamazepine-induced Stevens-Johnson syndrome. J. Allergy Clin. Immunol. 128:1277–78 [Google Scholar]
  54. Wei CY, Chung WH, Huang HW. 54.  et al. 2012. Direct interaction between HLA-B and carbamazepine activates T cells in patients with Stevens-Johnson syndrome. J. Allergy Clin. Immunol. 129:1562–69 [Google Scholar]
  55. Phillips E, Bartlett JA, Sanne I. 55.  et al. 2013. Associations between HLA-DRB1*0102, HLA-B*5801, and hepatotoxicity during initiation of nevirapine-containing regimens in South Africa. J. Acquir. Immune Defic. Syndr. 62:e55–57 [Google Scholar]
  56. Chantarangsu S, Mushiroda T, Mahasirimongkol S. 56.  et al. 2009. HLA-B*3505 allele is a strong predictor for nevirapine-induced skin adverse drug reactions in HIV-infected Thai patients. Pharmacogenet. Genomics 19:139–46 [Google Scholar]
  57. Gatanaga H, Yazaki H, Tanuma J. 57.  et al. 2007. HLA-Cw8 primarily associated with hypersensitivity to nevirapine. AIDS 21:264–65 [Google Scholar]
  58. Littera R, Carcassi C, Masala A. 58.  et al. 2006. HLA-dependent hypersensitivity to nevirapine in Sardinian HIV patients. AIDS 20:1621–26 [Google Scholar]
  59. Carr DF, Chaponda M, Jorgensen AL. 59.  et al. 2013. Association of human leukocyte antigen alleles and nevirapine hypersensitivity in a Malawian HIV-infected population. Clin. Infect. Dis. 56:1330–39 [Google Scholar]
  60. Gao S, Gui XE, Liang K. 60.  et al. 2012. HLA-dependent hypersensitivity reaction to nevirapine in Chinese Han HIV-infected patients. AIDS Res. Hum. Retroviruses 28:540–43 [Google Scholar]
  61. Likanonsakul S, Rattanatham T, Feangvad S. 61.  et al. 2009. HLA-Cw*04 allele associated with nevirapine-induced rash in HIV-infected Thai patients. AIDS Res. Ther. 6:22 [Google Scholar]
  62. Keane NM, Pavlos RK, McKinnon E. 62.  et al. 2014. HLA class I restricted CD8+ and class II restricted CD4+ T cells are implicated in the pathogenesis of nevirapine hypersensitivity. AIDS 28:1891–901 [Google Scholar]
  63. Persaud D, Gay H, Ziemniak C. 63.  et al. 2013. Absence of detectable HIV-1 viremia after treatment cessation in an infant. N. Engl. J. Med. 369:1828–35 [Google Scholar]
  64. Persaud D, Deveikis A, Gay H. 64.  et al. 2014. Very early combination antiretroviral therapy in perinatal HIV infection: two case studies Presented at Conf. Retroviruses and Opportunistic Infections, Boston
  65. Ciccacci C, Di Fusco D, Marazzi MC. 65.  et al. 2013. Association between CYP2B6 polymorphisms and nevirapine-induced SJS/TEN: a pharmacogenetics study. Eur. J. Clin. Pharmacol. 69:1909–16 [Google Scholar]
  66. Dickinson L, Chaponda M, Carr DF. 66.  et al. 2014. Population pharmacokinetic and pharmacogenetic analysis of nevirapine in hypersensitive and tolerant HIV-infected patients from Malawi. Antimicrob. Agents Chemother. 58:706–12 [Google Scholar]
  67. Lehr T, Yuan J, Hall D. 67.  et al. 2011. Integration of absorption, distribution, metabolism, and elimination genotyping data into a population pharmacokinetic analysis of nevirapine. Pharmacogenet. Genomics 21:721–30 [Google Scholar]
  68. Lucena MI, Molokhia M, Shen Y. 68.  et al. 2011. Susceptibility to amoxicillin-clavulanate-induced liver injury is influenced by multiple HLA class I and II alleles. Gastroenterology 141:338–47 [Google Scholar]
  69. Stevens R, Coates E, Street J. 69.  et al. 2013. Comprehensive evaluation of two HLA-B17 monoclonal antibodies for flow cytometry-based HLA-B57/B58 screening prior to abacavir prescription. Int. J. Immunogenet. 40:311–15 [Google Scholar]
  70. Kaniwa N, Saito Y, Aihara M. 70.  et al. 2008. HLA-B locus in Japanese patients with anti-epileptics and allopurinol-related Stevens-Johnson syndrome and toxic epidermal necrolysis. Pharmacogenomics 9:1617–22 [Google Scholar]
  71. Tassaneeyakul W, Jantararoungtong T, Chen P. 71.  et al. 2009. Strong association between HLA-B*5801 and allopurinol-induced Stevens-Johnson syndrome and toxic epidermal necrolysis in a Thai population. Pharmacogenet. Genomics 19:704–9 [Google Scholar]
  72. Emmerson BT, Hazelton RA, Frazer IH. 72.  1988. Some adverse reactions to allopurinol may be mediated by lymphocyte reactivity to oxypurinol. Arthritis Rheum. 31:436–40 [Google Scholar]
  73. Yun J, Mattsson J, Schnyder K. 73.  et al. 2013. Allopurinol hypersensitivity is primarily mediated by dose-dependent oxypurinol-specific T cell response. Clin. Exp. Allergy 43:1246–55 [Google Scholar]
  74. Day RO, Graham GG, Hicks M. 74.  et al. 2007. Clinical pharmacokinetics and pharmacodynamics of allo-purinol and oxypurinol. Clin. Pharmacokinet. 46:623–44 [Google Scholar]
  75. Braden GL, Warzynski MJ, Golightly M, Ballow M. 75.  1994. Cell-mediated immunity in allopurinol-induced hypersensitivity. Clin. Immunol. Immunopathol. 70:145–51 [Google Scholar]
  76. Narzi D, Becker CM, Fiorillo MT. 76.  et al. 2012. Dynamical characterization of two differentially disease associated MHC class I proteins in complex with viral and self-peptides. J. Mol. Biol. 415:429–42 [Google Scholar]
  77. Yun J, Marcaida MJ, Eriksson KK. 77.  et al. 2014. Oxypurinol directly and immediately activates the drug-specific T cells via the preferential use of HLA-B*58:01. J. Immunol. 192:2984–93 [Google Scholar]
  78. White K, Gaudieri S, Phillips E. 78.  2013. HLA and pharmacogenomics of drug hypersensitivity. Handbook of Pharmagenomics and Stratified Medicine S Padmanabhan 1437–59 London: Academic, Elsevier [Google Scholar]
  79. Chung WH, Chang WC, Lee YS. 79.  et al. 2014. Genetic variants associated with phenytoin-related severe cutaneous adverse reactions. JAMA 312:525–34 [Google Scholar]
  80. Chung WH, Chang WC, Stocker SL. 80.  et al. 2014. Insights into the poor prognosis of allopurinol-induced severe cutaneous adverse reactions: the impact of renal insufficiency, high plasma levels of oxypurinol and granulysin. Ann. Rheum. Dis. In press
/content/journals/10.1146/annurev-med-050913-022745
Loading
/content/journals/10.1146/annurev-med-050913-022745
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error