References


Phillips EJ. Classifying ADRs - does dose matter?  Br J Clin Pharmacol. 2016; 81:10-2.

  1. Drug hypersensitivity. Pichler WJ (ed), Karger, Basel, 2007.
  2. Nuesch R, Srasuebkul P, Ananworanich J, et al. Monitoring the toxicity of antiretroviral therapy in resource limited settings: a prospective clinical trial cohort in Thailand. J Antimicrob Chemother. 2006; 58:637-44.
  3. Pirmohamed M, Park BK. HIV and drug allergy. Curr Opin Allergy Clin Immunol. 2001; 1:311-6.
  4. Borras-Blasco J, Navarro-Ruiz A, Borras C, Castera E. Adverse cutaneous reactions associated with the newest antiretroviral drugs in patients with human immunodeficiency virus infection. J Antimicrob Chemother. 2008; 62:879-888.
  5. Ward HA, Russo GG, Shrum J. Cutaneous manifestations of antiretroviral therapy. J Am Acad Dermatol. 2002; 46:284-293.
  6. Naranjo CA, Busto U, Sellers EM, et al. A method for estimating the probability of adverse drug reactions. Clin Pharmacol Ther. 1981; 30:239-245.
  7. Mallal S, Phillips E, Carosi G, et al. HLA-B*5701 screening for hypersensitivity to abacavir. New EngJ Med. 2008; 358:568-579.
  8. Keane NM, Pavlos RK, McKinnon E, et al. HLA Class I restricted CD8+ and Class II restricted CD4+ T cells are implicated in the pathogenesis of nevirapine hypersensitivity. AIDS 2014; 28:1891-1901.
  9. Cherry CL, Crowe SM. Zidovudine. In: Grayson ML et al (eds). Kucers' The Use of Antibiotics. 7th ed., chapter 225: CRC Press; 2017.
  10. Venter WD, Sanne I. Stavudine. In: Grayson ML et al (eds). Kucers' The Use of Antibiotics. 7th ed., chapter 229: CRC Press; 2017.
  11. Audsley J, Giles M, Lewin S. Lamivudine. In: Grayson ML et al (eds). Kucers' The Use of Antibiotics. 7th ed., chapter 228: CRC Press; 2017.
  12. Trevillyan J, Wright E. Emtricitabine. In: Grayson ML et al (eds). Kucers' The Use of Antibiotics. 7th ed., chapter 231: CRC Press; 2017.
  13. Nelson M, Schiavone M. Emtricitabine (FTC) for the treatment of HIV infection. Int J Clin Pract. 2004; 58:504-510.
  14. Nelson MR, Katlama C, Montaner JS, et al. The safety of tenofovir disoproxil fumarate for the treatment of HIV infection in adults: the first 4 years. AIDS 2007; 21:1273-1281.
  15. Pinto AN, Cooper DA. Tenofovir. In: Grayson ML et al (eds). Kucers' The Use of Antibiotics. 7th ed., chapter 232: CRC Press; 2017.
  16. Sax PE, Wohl D, Yin MT, et al. Tenofovir alafenamide versus tenofovir disoproxil fumarate, coformulated with elvitegravir, cobicistat, and emtricitabine, for initial treatment of HIV-1 infection: two randomised, double-blind, phase 3, non-inferiority trials. Lancet 2015; 385:2606-2615.
  17. Cameron PU, Trubiano JA. Abacavir. In: Grayson ML et al (eds). Kucers' The Use of Antibiotics. 7th ed., Chapter 230: CRC Press; 2017.
  18. Street AC, Thevarajan I. Efavirenz. In: Grayson ML et al (eds). Kucers' The Use of Antibiotics. 7th ed., chapter 236: CRC Press; 2017.
  19. Pozniak AL, Morales-Ramirez J, Katabira E, et al. Efficacy and safety of TMC278 in antiretroviral-naive HIV-1 patients: week 96 results of a phase IIb randomized trial. AIDS 2010; 24:55-65.
  20. Havlir D, Cheeseman SH, McLaughlin M, et al. High-dose nevirapine: safety, pharmacokinetics, and antiviral effect in patients with human immunodeficiency virus infection. J Infect Dis. 1995; 171:537-545.
  21. Morlese JF, Qazi NA, Gazzard BG, Nelson MR. Nevirapine-induced neuropsychiatric complications, a class effect of non-nucleoside reverse transcriptase inhibitors? AIDS 2002; 16:1840-1841.
  22. Pett SL, Emery S. Atazanavir. In: Grayson ML et al (eds). Kucers' The Use of Antibiotics. 7th ed., chapter 246: CRC Press; 2017.
  23. Torti C, Lapadula G, Antinori A, et al. Hyperbilirubinemia during atazanavir treatment in 2,404 patients in the Italian atazanavir expanded access program and MASTER Cohorts. Infection 2009; 37:244-249.
  24. Cheng AC, Muttucumaru R, Crowe SM. Darunavir. In: Grayson ML et al (eds). Kucers' The Use of Antibiotics. 7th ed., chapter 245: CRC Press; 2017.
  25. Tseng A, Hughes CA, Wu J, et al. Cobicistat versus ritonavir: similar pharmacokinetic enhancers but some important differences. Ann Pharmacother. 2017; 51:1008-1022.
  26. Monteiro P, Perez I, Pich J, et al. Creatine kinase elevation in HIV-1-infected patients receiving raltegravir-containing antiretroviral therapy: a cohort study. J Antimicrob Chemother. 2013; 68:404-408.
  27. Hoffmann C, Llibre JM. Neuropsychiatric adverse events with dolutegravir and other integrase strand transfer inhibitors. AIDS Rev. 2019; 21:4-10.
  28. Llibre JM, Montoliu A, Miro JM, et al. Discontinuation of dolutegravir, elvitegravir/cobicistat and raltegravir because of toxicity in a prospective cohort. HIV Med. 2019; 20:237-247.
  29. Eckard AR, McComsey GA. Weight gain and integrase inhibitors. Curr Opin Infect Dis. 2020; 33:10-19.
  30. Mallal S, Nolan D, Witt C, et al. Association between presence of HLA-B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir. Lancet 2002; 359:727-732.
  31. Lee FJ, Carr A. Tolerability of HIV integrase inhibitors. Curr Opin HIV AIDS. 2012; 7:422-428.
  32. Perry ME, Almaani N, Desai N, et al. Raltegravir-induced drug reaction with eosinophilia and systemic symptoms (DRESS) syndrome - implications for clinical practice and patient safety. Int J STD AIDS 2013; 24:639-642.
  33. Scaggs FA, Aziz MS, Palmisano EL, et al. Raltegravir-induced drug reaction with eosinophilia and systemic symptoms syndrome in a child. Annals Allergy Asthma Immunol 2016; 117:719-721.
  34. Thomas M, Hopkins C, Duffy E, et al. Association of the HLA-B*53:01 allele with drug reaction with eosinophilia and systemic symptoms (DRESS) syndrome during treatment of HIV infection with raltegravir. Clin Infect Dis. 2017; 64:1198-1203.
  35. McKoy JM, Bennett CL, Scheetz MH, et al. Hepatotoxicity associated with long- versus short-course HIV-prophylactic nevirapine use: a systematic review and meta-analysis from the Research on Adverse Drug events and Reports (RADAR) project. Drug Saf. 2009; 32:147-158.
  36. Coopman SA, Johnson RA, Platt R, Stern RS. Cutaneous disease and drug reactions in HIV infection. New Eng J Med. 1993; 328:1670-1674.
  37. Milpied-Homsi B, Moran EM, Phillips EJ. Antiviral drug allergy. Immunology and Allergy Clinics of North America 2014; 34:645-662.
  38. Isaacs T, Ngwanya MR, Dlamini S, Lehloenya RJ. Annular erythema and photosensitivity as manifestations of efavirenz-induced cutaneous reactions: a review of five consecutive cases. J Antimicrob Chemother. 2013; 68:2871-2874.
  39. Lehloenya RJ, Kgokolo M. Clinical presentations of severe cutaneous drug reactions in HIV-infected Africans. Dermatologic Clinics 2014; 32:227-235.
  40. Urbancic KF, Ierino F, Phillips E, et al. Taking the challenge: A protocolized approach to optimize Pneumocystis pneumonia prophylaxis in renal transplant recipients. Am J Transplant. 2018; 18:462-466.
  41. Krantz M, Stone CA, Abreo A, Phillips EJ. Oral challenge with trimethoprim-sulfamethoxazole in patients with “sulfa” antibiotic allergy referred to an outpatient drug allergy clinic. J Allergy Clin Immunol. 2019; 143:AB209.
  42. Lin D, Li WK, Rieder MJ. Cotrimoxazole for prophylaxis or treatment of opportunistic infections of HIV/AIDS in patients with previous history of hypersensitivity to cotrimoxazole. Cochrane Database Syst Rev. 2007; 2:CD005646.
  43. Yoshizawa S, Yasuoka A, Kikuchi Y, et al. A 5-day course of oral desensitization to trimethoprim/sulfamethoxazole (T/S) in patients with human immunodeficiency virus type-1 infection who were previously intolerant to T/S. Annals Allergy Asthma Immunol 2000; 85:241-244.
  44. Gompels MM, Simpson N, Snow M, et al. Desensitization to co-trimoxazole (trimethoprim-sulphamethoxazole) in HIV-infected patients: is patch testing a useful predictor of reaction? Journal Infection. 1999; 38:111-115.
  45. Gluckstein D, Ruskin J. Rapid oral desensitization to trimethoprim-sulfamethoxazole (TMP-SMZ): use in prophylaxis for Pneumocystis carinii pneumonia in patients with AIDS who were previously intolerant to TMP-SMZ. Clin Infect Dis. 1995; 20:849-853.
  46. Absar N, Daneshvar H, Beall G. Desensitization to trimethoprim/sulfamethoxazole in HIV-infected patients. J Allergy Clin Immunol. 1994; 93:1001-1005.
  47. Strom BL, Schinnar R, Apter AJ, et al. Absence of cross-reactivity between sulfonamide antibiotics and sulfonamide nonantibiotics. New Eng J Med. 2003; 349:1628-1635.
  48. Holtzer CD, Flaherty JF, Jr., Coleman RL. Cross-reactivity in HIV-infected patients switched from trimethoprim-sulfamethoxazole to dapsone. Pharmacotherapy 1998; 18:831-835.
  49. Jorde UP, Horowitz HW, Wormser GP. Utility of dapsone for prophylaxis of Pneumocystis carinii pneumonia in trimethoprim-sulfamethoxazole-intolerant, HIV-infected individuals. AIDS 1993; 7:355-359.
  50. Beumont MG, Graziani A, Ubel PA, MacGregor RR. Safety of dapsone as Pneumocystis carinii pneumonia prophylaxis in human immunodeficiency virus-infected patients with allergy to trimethoprim/sulfamethoxazole. Am J Med. 1996; 100:611-616.
  51. Urbancic KF, Pisasale D, Wight J, Trubiano JA. Dapsone safety in haematology patients: pathways to optimising Pneumocystis jirovecii pneumonia prophylaxis in haematology malignancy and transplant recipients. Transpl Infect Dis. 2018; 20:e12968.
  52. May SM, Motosue MS, Park MA. Dapsone is often tolerated in HIV-infected patients with history of sulfonamide antibiotic intolerance. J Allergy Clin Immunol In Practice. 2017; 5:831-833.
  53. Trubiano JA, Adkinson NF, Phillips EJ. Penicillin Allergy Is Not Necessarily Forever. JAMA 2017; 318:82-83.
  54. Olteanu C, Shear NH, Dodiuk-Gad RP. Practical Approach to Diagnosis and Management of Cutaneous Adverse Drug Reactions. In: Shear N, Dodiuk-Gad R (eds), Advances in Diagnosis and Management of Cutaneous Adverse Drug Reactions, 2019. Adis, Singapore.
  55. Redwood AJ, Pavlos RK, White KD, Phillips EJ. HLAs: Key regulators of T-cell-mediated drug hypersensitivity. HLA 2018; 91:3-16.
  56. Martin AM, Nolan D, James I, et al. Predisposition to nevirapine hypersensitivity associated with HLA-DRB1*0101 and abrogated by low CD4 T-cell counts. AIDS 2005; 19:97-9.
  57. Lefebvre M, Walencik A, Allavena C, et al. Rate of DRESS syndrome with raltegravir and role of the HLA-B*53: 01 allele. J Acquir Immune Defic Syndr. 2020; 85:e77-e80.
  58. Clay PG. The abacavir hypersensitivity reaction: a review. Clinical Therapeutics. 2002; 24:1502-1514.
  59. Hetherington S, Hughes AR, Mosteller M, et al. Genetic variations in HLA-B region and hypersensitivity reactions to abacavir. Lancet 2002; 359:1121-1122.
  60. Rauch A, Nolan D, Thurnheer C, et al. Refining abacavir hypersensitivity diagnoses using a structured clinical assessment and genetic testing in the Swiss HIV Cohort Study. Antiviral Therapy. 2008; 13:1019-1028.
  61. Martin AM, Nolan D, Gaudieri S, et al. Predisposition to abacavir hypersensitivity conferred by HLA-B*5701 and a haplotypic Hsp70-Hom variant. Proc Natl Acad Sci U S A. 2004; 101:4180-4185.
  62. Martin AM, Almeida CA, Cameron P, et al. Immune responses to abacavir in antigen-presenting cells from hypersensitive patients. AIDS 2007; 21:1233-1244.
  63. Lucas A, Lucas M, Strhyn A, et al. Abacavir-reactive memory T cells are present in drug naive individuals. PloS One 2015; 10:e0117160.
  64. Phillips EJ, Sullivan JR, Knowles SR, Shear NH. Utility of patch testing in patients with hypersensitivity syndromes associated with abacavir. AIDS 2002; 16:2223-2225.
  65. Ostrov DA, Grant BJ, Pompeu YA, et al. Drug hypersensitivity caused by alteration of the MHC-presented self-peptide repertoire. Proc Natl Acad Sci U S A 2012; 109:9959-64.
  66. Illing PT, Vivian JP, Dudek NL, et al. Immune self-reactivity triggered by drug-modified HLA-peptide repertoire. Nature 2012; 486:554-558.
  67. Norcross MA, Luo S, Lu L, et al. Abacavir induces loading of novel self-peptides into HLA-B*57: 01: an autoimmune model for HLA-associated drug hypersensitivity. AIDS 2012; 26:F21-29.
  68. Cardone M, Garcia K, Tilahun ME, et al. A transgenic mouse model for HLA-B*57:01-linked abacavir drug tolerance and reactivity. J Clin Invest. 2018; 128:2819-2832.
  69. Phillips EJ, Mallal SA. Active suppression rather than ignorance: tolerance to abacavir-induced HLA-B*57:01 peptide repertoire alteration. J Clin Invest. 2018; 128:2746-2749.
  70. Pavlos R, Deshpande P, Chopra A et al. New genetic predictors for abacavir tolerance in HLA-B*57:01 positive individuals. Hum Immunol. 2020; 81:300-304.
  71. Pavlos R, Mallal S, Phillips E. HLA and pharmacogenetics of drug hypersensitivity. Pharmacogenomics 2012; 13:1285-1306.73. Vitezica ZG, Milpied B, Lonjou C, et al. HLA-DRB1*01 associated with cutaneous hypersensitivity induced by nevirapine and efavirenz. AIDS 2008; 22:540-541.
  72. Martin AM, Nolan D, James I, et al. Predisposition to nevirapine hypersensitivity associated with HLA-DRB1*0101 and abrogated by low CD4 T-cell counts. AIDS 2005; 19:97-99.
  73. Carr DF, Chaponda M, Jorgensen AL, et al. Association of human leukocyte antigen alleles and nevirapine hypersensitivity in a Malawian HIV-infected population. Clin Infect Dis. 2013; 56:1330-1339.
  74. Carr DF, Bourgeois S, Chaponda M, et al. Genome-wide association study of nevirapine hypersensitivity in a sub-Saharan African HIV-infected population. J Antimicrob Chemother. 2017; 72:1152-1162.
  75. Pavlos R, McKinnon EJ, Ostrov DA, et al. Shared peptide binding of HLA Class I and II alleles associate with cutaneous nevirapine hypersensitivity and identify novel risk alleles. Sci Rep. 2017; 7:8653.
  76. Fagot JP, Mockenhaupt M, Bouwes-Bavinck JN, et al. Nevirapine and the risk of Stevens-Johnson syndrome or toxic epidermal necrolysis. AIDS 2001; 15:1843-1848.
  77. Mittmann N, Knowles SR, Koo M, et al. Incidence of toxic epidermal necrolysis and Stevens-Johnson Syndrome in an HIV cohort: an observational, retrospective case series study. Am J Clin Dermatol. 2012; 13:49-54.
  78. Cornejo Castro EM, Carr DF, Jorgensen AL, et al. HLA-allelotype associations with nevirapine-induced hypersensitivity reactions and hepatotoxicity: a systematic review of the literature and meta-analysis. Pharmacogenet Genomics. 2015; 25:186-198.
  79. Lefebvre M, Walencik A, Allavena C, et al. Rate of DRESS srndrome with raltegravir and role of the HLA-B*53: 01 allele. J Acquir Immune Defic Syndr. 2020; 85:e77-e80.
  80. Blumenthal KG, Peter JG, Trubiano JA, Phillips EJ. Antibiotic allergy. Lancet 2019; 393:183-198.
  81. Joint Task Force on Practice Parameters; American Academy of Allergy, Asthma and Immunology; American College of Allergy, Asthma and Immunology; Joint Council of Allergy, Asthma and Immunology. Drug allergy: an updated practice parameter. Ann Allergy Asthma Immunol. 2010; 105:259-273.
  82. Phillips EJ, Bigliardi P, Bircher AJ, et al. Controversies in drug allergy: Testing for delayed reactions. J Allergy Clin Immunol. 2019; 143:66-73.
  83. Konvinse KC, Phillips EJ, White KD, Trubiano JA. Old dog begging for new tricks: current practices and future directions in the diagnosis of delayed antimicrobial hypersensitivity. Curr Opin Infect Dis. 2016; 29:561-576.
  84. Giorgini S, Martinelli C, Tognetti L, et al. Use of patch testing for the diagnosis of abacavir-related hypersensitivity reaction in HIV patients. Dermatologic therapy. 2011; 24:591-594.
  85. Phillips EJ, Wong GA, Kaul R, et al. Clinical and immunogenetic correlates of abacavir hypersensitivity. AIDS 2005; 19:979-981.
  86. Peter J, Choshi P, Lehloenya RJ. Drug hypersensitivity in HIV infection. Curr Opin Allergy Clin Immunol. 2019; 19:272-282.
  87. Shear NH, Milpied B, Bruynzeel DP, Phillips EJ. A review of drug patch testing and implications for HIV clinicians. AIDS 2008; 22:999-1007.
  88. Suarez-Lorenzo I, Castillo-Sainz R, Carden-Santana MA, Carrillo-Diaz T. Severe reaction to emtricitabine and lamiduvine: evidence of cross-reactivity. Contact Dermatitis. 2016; 74:253-254.
  89. Sousa MJ, Cadinha S, Mota M, et al. Hypersensitivity to antiretroviral drugs. European Ann Allergy Clin Immunol. 2018; 50:277-280.
  90. Lehloenya RJ, Peter JG, Copascu A, et al. Delabeling delayed drug hypersensitivity: how far can you safely go? J Allergy Clin Immunol Pract. 2020; 8:2878-2895.e6.
  91. Lehloenya RJ, Isaacs T, Nyika T, et al. Early high-dose intravenous corticosteroids rapidly arrest Stevens Johnson syndrome and drug reaction with eosinophilia and systemic symptoms recurrence on drug re-exposure. J Allergy Clin Immunol Pract. 2021; 9:582-584.e1.
  92. Thong BY, Chia FL, Tan SC, et al. A retrospective study on sequential desensitization-rechallenge for antituberculosis drug allergy. Asia Pac Allergy. 2014; 4:156-163.
  93. Siripassorn K, Ruxrungtham K, Manosuthi W. Successful drug desensitization in patients with delayed-type allergic reactions to anti-tuberculosis drugs. Int J Infect Dis. 2018; 68:61-68.
  94. groups AE. Therapeutic guidelines: antibiotic. Version 16. Therapeutic Guidelines Limited, Melbourne, 2019.