1
|
Cadot R, Gery P, Lenief V, Nicolas JF, Vocanson M, Tauber M. Exploring recent advances in drugs severe cutaneous adverse reactions immunopathology. Allergy 2024. [PMID: 39295209 DOI: 10.1111/all.16316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/13/2024] [Accepted: 09/03/2024] [Indexed: 09/21/2024]
Abstract
Severe cutaneous adverse reactions to drugs (SCARs) are rare but life-threatening delayed allergies. While they primarily affect the skin, they can also affect internal organs. Accordingly, they present with diverse clinical symptoms that vary not only between SCARs subtypes but also among patients. Despite the availability of topical and systemic treatments, these only address the symptoms and not the cause. To develop more effective therapies, it is necessary to elucidate the complexity of the pathophysiology of SCARs in relation to their severity. In line with the new type IV hypersensitivity reactions nomenclature proposed by the European Academy of Allergy and Clinical Immunology (EAACI), this review highlights the current insights into the intricate immune mechanisms engaged, the interplay between the culprit drug and genetic predisposition in drug presentation mechanisms, but also how external factors, such as viruses, are implicated in SCARs. Their relevance to the development of targeted medicine is also discussed.
Collapse
Affiliation(s)
- Romane Cadot
- CIRI-Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- CNRS, UMR 5308, Lyon, France
| | - Perrine Gery
- CIRI-Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- CNRS, UMR 5308, Lyon, France
| | - Vanina Lenief
- CIRI-Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- CNRS, UMR 5308, Lyon, France
| | - Jean-François Nicolas
- CIRI-Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- CNRS, UMR 5308, Lyon, France
- Allergology and Clinical Immunology Department, Lyon Sud University Hospital, Pierre Bénite, France
| | - Marc Vocanson
- CIRI-Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- CNRS, UMR 5308, Lyon, France
| | - Marie Tauber
- CIRI-Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- CNRS, UMR 5308, Lyon, France
- Allergology and Clinical Immunology Department, Lyon Sud University Hospital, Pierre Bénite, France
- Reference center for toxic bullous dermatitis and severe cutaneous adverse reactions, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
2
|
Lee WI, Lam L, Bacchi S, Jiang M, Inglis JM, Smith W, Hissaria P. Antibiotic prophylaxis in immunosuppressed patients - Missed opportunities from trimethoprim-sulfamethoxazole allergy label. World Allergy Organ J 2024; 17:100856. [PMID: 38235260 PMCID: PMC10793173 DOI: 10.1016/j.waojou.2023.100856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024] Open
Abstract
Trimethoprim-sulfamethoxazole (TMP-SMX) is a broad spectrum antibiotic in use for more than 50 years. It has an important indication as first line agent in the prophylaxis of opportunistic infections, particularly Pneumocystis jirovecii pneumonia (PJP), in immunosuppressed patients. For those who have a history of allergy or severe intolerance to TMP-SMX, pentamidine, dapsone or atovaquone may be substituted; however there is evidence that TMP-SMX offers superior coverage for PJP, toxoplasmosis, and nocardiosis. Compared to pentamidine, it has the added benefit of cost-effectiveness and self-administration as opposed to required hospital attendance for administration. Many patients who report a history of allergy or adverse reaction to TMP-SMX (or "sulfur allergy") will be found not to be allergic; and even those who are allergic may be able to be desensitized. The evaluation and, where appropriate, removal of TMP-SMX allergy label enables the use of TMP-SMX for prophylaxis against opportunistic infections. This is a cost-effective intervention to optimize antimicrobial prescribing and reduce the risk of opportunistic infections in immunosuppressed patients.
Collapse
Affiliation(s)
- Wei-I Lee
- Department of Immunology, The Canberra Hospital, Yamba Drive, Garran, ACT, 2605, Australia
- Australian National University, Canberra, ACT, 2601, Australia
| | - Lydia Lam
- University of Adelaide, Adelaide SA 5005, Australia
| | - Stephen Bacchi
- University of Adelaide, Adelaide SA 5005, Australia
- Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Melinda Jiang
- Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Joshua M. Inglis
- University of Adelaide, Adelaide SA 5005, Australia
- Flinders Medical Centre and University, Bedford Park, SA, 5042, Australia
| | - William Smith
- University of Adelaide, Adelaide SA 5005, Australia
- Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Pravin Hissaria
- University of Adelaide, Adelaide SA 5005, Australia
- Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Department of Immunopathology, SA Pathology, Frome Rd, Adelaide, 5000, Australia
| |
Collapse
|
3
|
Almutairi M, Lister A, Zhao Q, Line J, Adair K, Tailor A, Waddington J, Clarke E, Gardner J, Thomson P, Harper N, Sun Y, Sun L, Ostrov DA, Liu H, MacEwan DJ, Pirmohamed M, Meng X, Zhang F, Naisbitt DJ. Activation of Human CD8+ T Cells with Nitroso Dapsone-Modified HLA-B*13:01-Binding Peptides. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1031-1042. [PMID: 36881872 PMCID: PMC7614401 DOI: 10.4049/jimmunol.2200531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 02/11/2023] [Indexed: 03/09/2023]
Abstract
Previous studies have shown that cysteine-reactive drug metabolites bind covalently with protein to activate patient T cells. However, the nature of the antigenic determinants that interact with HLA and whether T cell stimulatory peptides contain the bound drug metabolite has not been defined. Because susceptibility to dapsone hypersensitivity is associated with the expression of HLA-B*13:01, we have designed and synthesized nitroso dapsone-modified, HLA-B*13:01 binding peptides and explored their immunogenicity using T cells from hypersensitive human patients. Cysteine-containing 9-mer peptides with high binding affinity to HLA-B*13:01 were designed (AQDCEAAAL [Pep1], AQDACEAAL [Pep2], and AQDAEACAL [Pep3]), and the cysteine residue was modified with nitroso dapsone. CD8+ T cell clones were generated and characterized in terms of phenotype, function, and cross-reactivity. Autologous APCs and C1R cells expressing HLA-B*13:01 were used to determine HLA restriction. Mass spectrometry confirmed that nitroso dapsone-peptides were modified at the appropriate site and were free of soluble dapsone and nitroso dapsone. APC HLA-B*13:01-restricted nitroso dapsone-modified Pep1- (n = 124) and Pep3-responsive (n = 48) CD8+ clones were generated. Clones proliferated and secreted effector molecules with graded concentrations of nitroso dapsone-modified Pep1 or Pep3. They also displayed reactivity against soluble nitroso dapsone, which forms adducts in situ, but not with the unmodified peptide or dapsone. Cross-reactivity was observed between nitroso dapsone-modified peptides with cysteine residues in different positions in the peptide sequence. These data characterize a drug metabolite hapten CD8+ T cell response in an HLA risk allele-restricted form of drug hypersensitivity and provide a framework for structural analysis of hapten HLA binding interactions.
Collapse
Affiliation(s)
- Mubarak Almutairi
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Adam Lister
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Qing Zhao
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - James Line
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Kareena Adair
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Arun Tailor
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - James Waddington
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Elsie Clarke
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Joshua Gardner
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Paul Thomson
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Nicolas Harper
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Yonghu Sun
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lele Sun
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - David A. Ostrov
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Hong Liu
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - David J. MacEwan
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Munir Pirmohamed
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Furen Zhang
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Dean J Naisbitt
- MRC Centre for Drug Safety Science, Dept. Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| |
Collapse
|
4
|
Pezeshki PS, Nowroozi A, Razi S, Rezaei N. Asthma and Allergy. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
5
|
Juncos LA, Wieruszewski PM, Kashani K. Pathophysiology of Acute Kidney Injury in Critical Illness: A Narrative Review. Compr Physiol 2022; 12:3767-3780. [PMID: 36073750 DOI: 10.1002/cphy.c210028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Acute kidney injury (AKI) is a syndrome that entails a rapid decline in kidney function with or without injury. The consequences of AKI among acutely ill patients are dire and lead to higher mortality, morbidity, and healthcare cost. To prevent AKI and its short and long-term repercussions, understanding its pathophysiology is essential. Depending on the baseline kidney histology and function reserves, the number of kidney insults, and the intensity of each insult, the clinical presentation of AKI may differ. While many factors are capable of inducing renal injury, they can be categorized into a few processes. The three primary processes reported in the literature are hemodynamic changes, inflammatory reactions, and nephrotoxicity. The majority of patients with AKI will suffer from more than one during their development and/or progression of AKI. Moreover, the development of one usually leads to the instigation of another. Thus, the interactions and progression between these mechanisms may determine the severity and duration of the AKI. Other factors such as organ crosstalk and how our concurrent therapies interact with these mechanisms complicate the pathophysiology of the progression of the AKI even further. In this narrative review article, we describe these three main pathophysiological processes that lead to the development and progression of AKI. © 2022 American Physiological Society. Compr Physiol 12: 1-14, 2022.
Collapse
Affiliation(s)
- Luis A Juncos
- Division of Nephrology, Central Arkansas Veterans' Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Patrick M Wieruszewski
- Division of Hospital Pharmacy, Department of Pharmacy, Mayo Clinic, Rochester, Minnesota, USA
| | - Kianoush Kashani
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
6
|
Adair K, Meng X, Naisbitt DJ. Drug hapten-specific T-cell activation: Current status and unanswered questions. Proteomics 2021; 21:e2000267. [PMID: 33651918 DOI: 10.1002/pmic.202000267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/14/2021] [Accepted: 02/16/2021] [Indexed: 11/07/2022]
Abstract
Drug haptens are formed from the irreversible, covalent binding of drugs to nucleophilic moieties on proteins, which can warrant adverse reactions in the body including severe delayed-type, T-cell mediated, drug hypersensitivity reactions (DHRs). While three main pathways exist for the activation of T-cells in DHRs, namely the hapten model, the pharmacological interaction model and the altered peptide repertoire model, the exact antigenic determinants responsible have not yet been defined. In recent years, progress has been made using advanced mass spectrometry-based proteomic methods to identify protein carriers and characterise the structure of drug-haptenated proteins. Since genome-wide association studies discovered a link between human leukocyte antigens (HLA) and an individual's susceptibility to DHRs, much effort has been made to define the drug-associated HLA ligands driving T-cell activation, including the elution of natural HLA peptides from HLA molecules and the generation of HLA-binding peptides. In this review, we discuss our current methodology used to design and synthesise drug-modified HLA ligands to investigate their immunogenicity using T-cell models, and thus their implication in drug hypersensitivity.
Collapse
Affiliation(s)
- Kareena Adair
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - Xiaoli Meng
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - Dean J Naisbitt
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| |
Collapse
|
7
|
Elzagallaai AA, Sultan EA, Bend JR, Abuzgaia AM, Loubani E, Rieder MJ. Role of Oxidative Stress in Hypersensitivity Reactions to Sulfonamides. J Clin Pharmacol 2019; 60:409-421. [PMID: 31709574 DOI: 10.1002/jcph.1535] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/26/2019] [Indexed: 12/16/2022]
Abstract
Antimicrobial sulfonamides are important medications. However, their use is associated with major immune-mediated drug hypersensitivity reactions with a rate that ranges from 3% to 4% in the general population. The pathophysiology of sulfa-induced drug hypersensitivity reactions is not well understood, but accumulation of reactive metabolites (sulfamethoxazole [SMX] hydroxylamine [SMX-HA] and SMX N-nitrosamine [SMX-NO]) is thought to be a major factor. These reactive metabolites contribute to the formation of reactive oxygen species (ROS) known to cause cellular damage and induce cell death through apoptosis and necroptosis. ROS can also serve as "danger signals," priming immune cells to mount an immunological reaction. We recruited 26 sulfa-hypersensitive (HS) patients, 19 healthy control subjects, and 6 sulfa-tolerant patients to this study. Peripheral blood monocytes and platelets were isolated from blood samples and analyzed for in vitro cytotoxicity, ROS and carbonyl protein formation, lipid peroxidation, and GSH (glutathione) content after challenge with SMX-HA. When challenged with SMX-HA, cells isolated from sulfa-HS patients exhibited significantly (P ≤ .05) higher cell death, ROS and carbonyl protein formation, and lipid peroxidation. In addition, there was a high correlation between cell death in PBMCs and ROS levels. There was also depletion of GSH and lower GSH/GSSG ratios in peripheral blood mononuclear cells from sulfa-HS patients. The amount of ROS formed was negatively correlated with intracellular GSH content. The data demonstrate a major role for oxidative stress in in vitro cytotoxicity of SMX reactive metabolites and indicate increased vulnerability of cells from sulfa-HS patients to the in vitro challenge.
Collapse
Affiliation(s)
- Abdelbaset A Elzagallaai
- Department of Paediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Elham A Sultan
- Department of Paediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - John R Bend
- Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Awatif M Abuzgaia
- Department of Paediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Eman Loubani
- Department of Paediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Michael J Rieder
- Department of Paediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
8
|
Mayorga C, Montañez MI, Jurado-Escobar R, Gil-Ocaña V, Cornejo-García JA. An Update on the Immunological, Metabolic and Genetic Mechanisms in Drug Hypersensitivity Reactions. Curr Pharm Des 2019; 25:3813-3828. [PMID: 31692430 DOI: 10.2174/1381612825666191105122414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/31/2019] [Indexed: 11/22/2022]
Abstract
Drug hypersensitivity reactions (DHRs) represent a major burden on the healthcare system since their diagnostic and management are complex. As they can be influenced by individual genetic background, it is conceivable that the identification of variants in genes potentially involved could be used in genetic testing for the prevention of adverse effects during drug administration. Most genetic studies on severe DHRs have documented HLA alleles as risk factors and some mechanistic models support these associations, which try to shed light on the interaction between drugs and the immune system during lymphocyte presentation. In this sense, drugs are small molecules that behave as haptens, and currently three hypotheses try to explain how they interact with the immune system to induce DHRs: the hapten hypothesis, the direct pharmacological interaction of drugs with immune receptors hypothesis (p-i concept), and the altered self-peptide repertoire hypothesis. The interaction will depend on the nature of the drug and its reactivity, the metabolites generated and the specific HLA alleles. However, there is still a need of a better understanding of the different aspects related to the immunological mechanism, the drug determinants that are finally presented as well as the genetic factors for increasing the risk of suffering DHRs. Most available information on the predictive capacity of genetic testing refers to abacavir hypersensitivity and anticonvulsants-induced severe cutaneous reactions. Better understanding of the underlying mechanisms of DHRs will help us to identify the drugs likely to induce DHRs and to manage patients at risk.
Collapse
Affiliation(s)
- Cristobalina Mayorga
- Allergy Research Group, Instituto de Investigacion Biomedica de Malaga-IBIMA-ARADyAL. Malaga, Spain.,Allergy Unit, Hospital Regional Universitario de Málaga-ARADyAL. Málaga, Spain.,Andalusian Center for Nanomedicine and Biotechnology-BIONAND. Malaga, Spain
| | - Maria I Montañez
- Allergy Research Group, Instituto de Investigacion Biomedica de Malaga-IBIMA-ARADyAL. Malaga, Spain.,Andalusian Center for Nanomedicine and Biotechnology-BIONAND. Malaga, Spain
| | - Raquel Jurado-Escobar
- Allergy Research Group, Instituto de Investigacion Biomedica de Malaga-IBIMA-ARADyAL. Malaga, Spain.,Universidad de Málaga, Málaga, Spain
| | - Violeta Gil-Ocaña
- Andalusian Center for Nanomedicine and Biotechnology-BIONAND. Malaga, Spain.,Department of Organic Chemistry, Universidad de Málaga, ARADyAL, Málaga, Spain
| | - Jose A Cornejo-García
- Allergy Research Group, Instituto de Investigacion Biomedica de Malaga-IBIMA-ARADyAL. Malaga, Spain
| |
Collapse
|
9
|
Tailor A, Waddington JC, Hamlett J, Maggs J, Kafu L, Farrell J, Dear GJ, Whitaker P, Naisbitt DJ, Park K, Meng X. Definition of Haptens Derived from Sulfamethoxazole: In Vitro and in Vivo. Chem Res Toxicol 2019; 32:2095-2106. [DOI: 10.1021/acs.chemrestox.9b00282] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Arun Tailor
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, U.K
| | - James C. Waddington
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, U.K
| | - Jane Hamlett
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, U.K
| | - James Maggs
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, U.K
| | - Laila Kafu
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, U.K
| | - John Farrell
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, U.K
| | - Gordon J. Dear
- GlaxoSmithKline, Park Road, Ware, Hertfordshire SG12 0DP, U.K
| | - Paul Whitaker
- Regional Adult Cystic Fibrosis Unit, St. James’s University Hospital, Leeds LS9 7TF, U.K
| | - Dean J. Naisbitt
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, U.K
| | - Kevin Park
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, U.K
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool L69 3GE, U.K
| |
Collapse
|
10
|
Dal Negro G, Eskes C, Belz S, Bertein C, Chlebus M, Corvaro M, Corvi R, Dhalluin S, Halder M, Harvey J, Hermann M, Hoffmann-Dörr S, Kilian K, Lambrigts D, Laroche C, Louhimies S, Mahony C, Manou I, McNamee P, Prieto P, Reid K, Roggen E, Schutte K, Stirling C, Uhlrich S, Weissenhorn R, Whelan M. One science-driven approach for the regulatory implementation of alternative methods: A multi-sector perspective. Regul Toxicol Pharmacol 2018; 99:33-49. [PMID: 30098372 DOI: 10.1016/j.yrtph.2018.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/05/2018] [Indexed: 12/28/2022]
Abstract
EU regulations call for the use of alternative methods to animal testing. During the last decade, an increasing number of alternative approaches have been formally adopted. In parallel, new 3Rs-relevant technologies and mechanistic approaches have increasingly contributed to hazard identification and risk assessment evolution. In this changing landscape, an EPAA meeting reviewed the challenges that different industry sectors face in the implementation of alternative methods following a science-driven approach. Although clear progress was acknowledged in animal testing reduction and refinement thanks to an integration of scientifically robust approaches, the following challenges were identified: i) further characterization of toxicity pathways; ii) development of assays covering current scientific gaps, iii) better characterization of links between in vitro readouts and outcome in the target species; iv) better definition of alternative method applicability domains, and v) appropriate implementation of the available approaches. For areas having regulatory adopted alternative methods (e.g., vaccine batch testing), harmonised acceptance across geographical regions was considered critical for broader application. Overall, the main constraints to the application of non-animal alternatives are the still existing gaps in scientific knowledge and technological limitations. The science-driven identification of most appropriate methods is key for furthering a multi-sectorial decrease in animal testing.
Collapse
Affiliation(s)
- Gianni Dal Negro
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, United Kingdom
| | - Chantra Eskes
- SeCAM Services and Consultation on Alternative Methods, Via Campagnora 1, 6983, Magliaso, Switzerland.
| | - Susanne Belz
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21017, Ispra, Italy
| | | | - Magda Chlebus
- European Federation of Pharmaceutical Industries and Associations (EFPIA), Rue du Trône 108, 1050, Brussels, Belgium
| | - Marco Corvaro
- ECPA - the European Crop Protection Association, 6 Avenue E. Van Nieuwenhuyse, 1160, Brussels, Belgium
| | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21017, Ispra, Italy
| | - Stephane Dhalluin
- L'Oréal Research & Innovation, 9 rue Pierre Dreyfus, 92110, Clichy, France
| | - Marlies Halder
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21017, Ispra, Italy
| | - Jim Harvey
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, United Kingdom
| | - Martina Hermann
- Henkel AG & Co. KGaA, Henkelstr. 67, 40589, Duesseldorf, Germany
| | | | - Karin Kilian
- European Commission, Directorate General for the Environment (DG ENV), Brussels, Belgium
| | - Denis Lambrigts
- GlaxoSmithKline Vaccines, 20 Avenue Fleming, 1300, Wavre, Belgium
| | - Charles Laroche
- European Partnership for Alternative Approaches to Animal Testing (EPAA), Av. Herrmann-Debroux 40, 1160, Brussels, Belgium
| | - Susanna Louhimies
- European Commission, Directorate General for the Environment (DG ENV), Brussels, Belgium
| | - Catherine Mahony
- The Procter & Gamble Company, Whitehall Lane, Egham, Surrey TW20 9NW, United Kingdom
| | - Irene Manou
- European Partnership for Alternative Approaches to Animal Testing (EPAA), Av. Herrmann-Debroux 40, 1160, Brussels, Belgium
| | - Pauline McNamee
- The Procter & Gamble Company, Whitehall Lane, Egham, Surrey TW20 9NW, United Kingdom
| | - Pilar Prieto
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21017, Ispra, Italy
| | - Kirsty Reid
- European Federation of Pharmaceutical Industries and Associations (EFPIA), Rue du Trône 108, 1050, Brussels, Belgium
| | - Erwin Roggen
- 3Rs Management and Consulting, Asavænget 14, 2800, Kongens Lyngby, Denmark
| | - Katrin Schutte
- European Commission, Directorate General for the Environment (DG ENV), Brussels, Belgium
| | | | - Sylvie Uhlrich
- Sanofi Pasteur, 1541 Av. Marcel Merieux, 69280, Marcy l'Etoile, France
| | - Renate Weissenhorn
- European Commission, Directorate General for Internal Market, Industry, Enterpreneurship and SME, Brussels, Belgium
| | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21017, Ispra, Italy
| |
Collapse
|
11
|
Adeyanju K, Bend JR, Rieder MJ, Dekaban GA. HIV-1 tat expression and sulphamethoxazole hydroxylamine mediated oxidative stress alter the disulfide proteome in Jurkat T cells. Virol J 2018; 15:82. [PMID: 29743079 PMCID: PMC5944096 DOI: 10.1186/s12985-018-0991-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/26/2018] [Indexed: 12/30/2022] Open
Abstract
Background Adverse drug reactions (ADRs) are a significant problem for HIV patients, with the risk of developing ADRs increasing as the infection progresses to AIDS. However, the pathophysiology underlying ADRs remains unknown. Sulphamethoxazole (SMX) via its active metabolite SMX-hydroxlyamine, when used prophylactically for pneumocystis pneumonia in HIV-positive individuals, is responsible for a high incidence of ADRs. We previously demonstrated that the HIV infection and, more specifically, that the HIV-1 Tat protein can exacerbate SMX-HA-mediated ADRs. In the current study, Jurkat T cell lines expressing Tat and its deletion mutants were used to determine the effect of Tat on the thiol proteome in the presence and absence of SMX-HA revealing drug-dependent changes in the disulfide proteome in HIV infected cells. Protein lysates from HIV infected Jurkat T cells and Jurkat T cells stably transfected with HIV Tat and Tat deletion mutants were subjected to quantitative slot blot analysis, western blot analysis and redox 2 dimensional (2D) gel electrophoresis to analyze the effects of SMX-HA on the thiol proteome. Results Redox 2D gel electrophoresis demonstrated that untreated, Tat-expressing cells contain a number of proteins with oxidized thiols. The most prominent of these protein thiols was identified as peroxiredoxin. The untreated, Tat-expressing cell lines had lower levels of peroxiredoxin compared to the parental Jurkat E6.1 T cell line. Conversely, incubation with SMX-HA led to a 2- to 3-fold increase in thiol protein oxidation as well as a significant reduction in the level of peroxiredoxin in all the cell lines, particularly in the Tat-expressing cell lines. Conclusion SMX-HA is an oxidant capable of inducing the oxidation of reactive protein cysteine thiols, the majority of which formed intermolecular protein bonds. The HIV Tat-expressing cell lines showed greater levels of oxidative stress than the Jurkat E6.1 cell line when treated with SMX-HA. Therefore, the combination of HIV Tat and SMX-HA appears to alter the activity of cellular proteins required for redox homeostasis and thereby accentuate the cytopathic effects associated with HIV infection of T cells that sets the stage for the initiation of an ADR. Electronic supplementary material The online version of this article (10.1186/s12985-018-0991-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kemi Adeyanju
- BioTherapeutics Research Laboratory, Molecular Medicine Research Laboratories, Robarts Research Institute, Rm 2214, 1151 Richmond Street North, London, Ontario, Canada.,Department of Microbiology and Immunology, University of Western Ontario, 1151 Richmond Street North, London, ON, N6A 5B7, Canada
| | - John R Bend
- Department of Pathology and Laboratory Medicine, University of Western Ontario, 1151 Richmond Street North, London, ON, N6A 5B7, Canada
| | - Michael J Rieder
- Drug Safety Laboratory, Molecular Medicine Research Laboratories, Robarts Research Institute, Rm 2214, 1151 Richmond Street North, London, Ontario, Canada.,Department of Pediatrics, University of Western Ontario, 1151 Richmond Street North, London, ON, N6A 5B7, Canada
| | - Gregory A Dekaban
- BioTherapeutics Research Laboratory, Molecular Medicine Research Laboratories, Robarts Research Institute, Rm 2214, 1151 Richmond Street North, London, Ontario, Canada. .,Department of Microbiology and Immunology, University of Western Ontario, 1151 Richmond Street North, London, ON, N6A 5B7, Canada.
| |
Collapse
|
12
|
Wong YY, Johnson B, Friedrich TC, Trepanier LA. Hepatic expression profiles in retroviral infection: relevance to drug hypersensitivity risk. Pharmacol Res Perspect 2017; 5:e00312. [PMID: 28603631 PMCID: PMC5464341 DOI: 10.1002/prp2.312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 12/11/2022] Open
Abstract
HIV‐infected patients show a markedly increased risk of delayed hypersensitivity (HS) reactions to potentiated sulfonamide antibiotics (trimethoprim/sulfamethoxazole or TMP/SMX). Some studies have suggested altered SMX biotransformation in HIV infection, but hepatic biotransformation pathways have not been evaluated directly. Systemic lupus erythematosus (SLE) is another chronic inflammatory disease with a higher incidence of sulfonamide HS, but it is unclear whether retroviral infection and SLE share risk factors for drug HS. We hypothesized that retroviral infection would lead to dysregulation of hepatic pathways of SMX biotransformation, as well as pathway alterations in common with SLE that could contribute to drug HS risk. We characterized hepatic expression profiles and enzymatic activities in an SIV‐infected macaque model of retroviral infection, and found no evidence for dysregulation of sulfonamide drug biotransformation pathways. Specifically, NAT1,NAT2,CYP2C8,CYP2C9,CYB5R3,MARC1/2, and glutathione‐related genes (GCLC,GCLM,GSS,GSTM1, and GSTP1) were not differentially expressed in drug naïve SIVmac239‐infected male macaques compared to age‐matched controls, and activities for SMX N‐acetylation and SMX hydroxylamine reduction were not different. However, multiple genes that are reportedly over‐expressed in SLE patients were also up‐regulated in retroviral infection, to include enhanced immunoproteasomal processing and presentation of antigens as well as up‐regulation of gene clusters that may be permissive to autoimmunity. These findings support the hypothesis that pathways downstream from drug biotransformation may be primarily important in drug HS risk in HIV infection.
Collapse
Affiliation(s)
- Yat Yee Wong
- Department of Medical Sciences School of Veterinary Medicine Madison Wisconsin
| | - Brian Johnson
- Molecular and Environmental Toxicology Center School of Medicine and Public Health University of Wisconsin-Madison Madison Wisconsin
| | - Thomas C Friedrich
- Department of Pathobiological Sciences School of Veterinary Medicine Madison Wisconsin.,AIDS Vaccine Research Laboratory Wisconsin National Primate Research Center Madison Wisconsin
| | - Lauren A Trepanier
- Department of Medical Sciences School of Veterinary Medicine Madison Wisconsin
| |
Collapse
|
13
|
Raghavan R, Shawar S. Mechanisms of Drug-Induced Interstitial Nephritis. Adv Chronic Kidney Dis 2017; 24:64-71. [PMID: 28284381 DOI: 10.1053/j.ackd.2016.11.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/01/2016] [Accepted: 11/03/2016] [Indexed: 11/11/2022]
Abstract
Drug-induced acute interstitial nephritis (DI-AIN) is a drug hypersensitivity reaction (DHR) that manifests 7 to 10 days after exposure to the culprit drug. DHRs account for fewer than 15% of reported adverse drug reactions. The kidneys are susceptible to DHR because: (1) the high renal blood flow whereby antigens are filtered, secreted, or concentrated, and (2) it is a major site of excretion for drugs and drug metabolites. More than 250 different drugs from various classes have been incriminated as causative agents of DI-AIN, the third most common cause of acute kidney injury in the hospital. DI-AIN must be differentiated from drug-induced nephrotoxic acute tubular necrosis because of their differing pathophysiology and treatment. DI-AIN begins with antigen processing and presentation to local dendritic cells. The dendritic cells activate T cells, and the subsequent effector phase of the immune response is mediated by various cytokines. Incriminated antigenic mechanisms include response to a conjugation product of the drug or its metabolite with a host protein (eg, beta-lactam or sulfonamide antibiotic) or the direct binding of the drug to a particular host allele to elicit a hypersensitivity response (eg, certain anti-epileptic drugs). If the offending drug is not identified and discontinued in a timely manner, irreversible fibrosis and chronic kidney disease will occur. The core structure of each drug or its metabolite is an antigenic determinant, and the host interaction is termed the structure-activity relationship. Differing structure-activity relationships accounts for effect, hypersensitivity, and cross-reactivity among and between classes. The essence of management of DI-AIN lies with the four sequential steps: anticipation, diagnosis, treatment, and prevention. Corticosteroids are used in the treatment of DI-AIN because of their potent anti-inflammatory effects on T cells and eosinophils. Anticipation and prevention require notifying the patient that DI-AIN is an idiosyncratic, hypersensitivity reaction that recurs on re-exposure, and the drug should be avoided.
Collapse
|
14
|
Tailor A, Waddington JC, Meng X, Park BK. Mass Spectrometric and Functional Aspects of Drug–Protein Conjugation. Chem Res Toxicol 2016; 29:1912-1935. [DOI: 10.1021/acs.chemrestox.6b00147] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Arun Tailor
- MRC Center
for Drug Safety
Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - James C. Waddington
- MRC Center
for Drug Safety
Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - Xiaoli Meng
- MRC Center
for Drug Safety
Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - B. Kevin Park
- MRC Center
for Drug Safety
Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, United Kingdom
| |
Collapse
|
15
|
Wong YY, Rakasz EG, Gasper DJ, Friedrich TC, Trepanier LA. Immunogenicity of trimethoprim/sulfamethoxazole in a macaque model of HIV infection. Toxicology 2016; 368-369:10-18. [PMID: 27565715 DOI: 10.1016/j.tox.2016.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 08/08/2016] [Accepted: 08/09/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Sulfonamide hypersensitivity has a high incidence in HIV infection and correlates with low CD4+ counts, but the mechanisms are not understood. The aims of this study were to determine whether trimethoprim/sulfamethoxazole (TMP/SMX) led to SMX adduct formation, immunogenicity, or signs of drug hypersensitivity in SIV-infected rhesus macaques, and whether differences in antioxidants, pro-inflammatory mediators, or SMX disposition were predictive of drug immunogenicity. METHODS Nine macaques chronically infected with SIVmac239 and 7 non-infected controls were studied. Baseline blood ascorbate, glutathione, IFN-γ, LPS, sCD14, and cytochrome b5 reductase measurements were obtained, macaques were dosed with TMP/SMX (120mg/kg/day p.o. for 14days), and SMX metabolites, lymph node drug adducts, drug-responsive T cells, and anti-SMX antibodies were measured. RESULTS Four of 9 of SIV-positive (44%), and 3 of 7 SIV negative (43%) macaques had drug-responsive T cells or antibodies to SMX. Two macaques developed facial or truncal rash; these animals had the highest levels of lymph node drug adducts. Antioxidants, pro-inflammatory mediators, and SMX metabolites were not predictive of drug immunogenicity; however, the Mamu DRB1*0401/0406/0411 genotype was significantly over-represented in immune responders. CONCLUSIONS Unlike other animal models, macaques develop an immune response, and possible rash, in response to therapeutic dosages of TMP/SMX. Studying more animals with CD4+ counts <200cells/μl, along with moderately restricted ascorbate intake to match deficiencies seen in humans, may better model the risk of SMX hypersensitivity in HIV-infection. In addition, the role of Mamu-DRB1 genotype in modeling drug hypersensitivity in retroviral infection deserves further study.
Collapse
Affiliation(s)
- Yat Yee Wong
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Eva G Rakasz
- AIDS Vaccine Research Laboratory, Wisconsin National Primate Research Center, Madison, WI, USA
| | - David J Gasper
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Thomas C Friedrich
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA; AIDS Vaccine Research Laboratory, Wisconsin National Primate Research Center, Madison, WI, USA
| | - Lauren A Trepanier
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
16
|
Goldman JL, Leeder JS, Van Haandel L, Pearce RE. In Vitro Hepatic Oxidative Biotransformation of Trimethoprim. Drug Metab Dispos 2015; 43:1372-80. [PMID: 26138612 DOI: 10.1124/dmd.115.065193] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/02/2015] [Indexed: 12/16/2022] Open
Abstract
Trimethoprim (TMP) has been widely used since the 1960s, both alone and in combination with sulfamethoxazole. Unfortunately, information regarding the role that cytochrome P450 enzymes (P450s) play in the formation of TMP primary metabolites is scarce. Hence, we undertook in vitro studies to identify and more fully characterize the P450s that catalyze formation of six TMP primary metabolites: TMP 1-N-oxide (1-NO-TMP) and 3-N-oxide (3-NO-TMP), 3'- and 4'-desmethyl-TMP, a benzylic alcohol (Cα-OH-TMP), and an N-acetyl cysteine (NAC) adduct of TMP (Cα-NAC-TMP). Formation kinetics for each TMP metabolite in human liver microsomes (HLMs) were consistent with single-enzyme Michaelis-Menten kinetics, and Km values were markedly above (≥10-fold) the therapeutic concentrations of TMP (50 µM). The combined results from correlation studies between rates of metabolite formation and marker P450 activities in a panel of HLMs along with inhibition studies utilizing selective P450 inhibitors incubated with pooled HLMs suggested that 1-NO-TMP, Cα-NAC-TMP, and Cα-OH-TMP were predominantly formed by CYP3A4. In contrast, 3-NO-TMP was formed predominantly by CYP1A2 in HLMs and inhibited by α-naphthoflavone. 4'-Desmethyl-TMP, which is believed to be a reactive TMP metabolite precursor, was formed by several P450s, including CYP3A4, correlated with multiple P450 activities, but was inhibited primarily by ketoconazole (up to 50%), suggesting that CYP3A4 makes a major contribution to TMP 4'-demethylation. TMP 3'-demethylation was catalyzed by multiple P450s, including CYP2C9, correlated with CYP2C9 activity, and was inhibited by sulfaphenazole (up to 40%). Overall, CYP2C9 and CYP3A4 appear to be the most significant contributors to TMP primary metabolism.
Collapse
Affiliation(s)
- Jennifer L Goldman
- Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (J.L.G., J.S.L., L.V.H., R.E.P.) and Infectious Diseases (J.L.G.), Departments of Pediatrics (J.L.G., J.S.L., L.V.H., R.E.P.) and Pharmacology (J.S.L. R.E.P.), Children's Mercy Hospital, University of Missouri, Kansas City, Missouri
| | - J Steven Leeder
- Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (J.L.G., J.S.L., L.V.H., R.E.P.) and Infectious Diseases (J.L.G.), Departments of Pediatrics (J.L.G., J.S.L., L.V.H., R.E.P.) and Pharmacology (J.S.L. R.E.P.), Children's Mercy Hospital, University of Missouri, Kansas City, Missouri
| | - Leon Van Haandel
- Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (J.L.G., J.S.L., L.V.H., R.E.P.) and Infectious Diseases (J.L.G.), Departments of Pediatrics (J.L.G., J.S.L., L.V.H., R.E.P.) and Pharmacology (J.S.L. R.E.P.), Children's Mercy Hospital, University of Missouri, Kansas City, Missouri
| | - Robin E Pearce
- Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (J.L.G., J.S.L., L.V.H., R.E.P.) and Infectious Diseases (J.L.G.), Departments of Pediatrics (J.L.G., J.S.L., L.V.H., R.E.P.) and Pharmacology (J.S.L. R.E.P.), Children's Mercy Hospital, University of Missouri, Kansas City, Missouri
| |
Collapse
|
17
|
Naisbitt DJ, Nattrass RG, Ogese MO. In vitro diagnosis of delayed-type drug hypersensitivity: mechanistic aspects and unmet needs. Immunol Allergy Clin North Am 2015; 34:691-705, x. [PMID: 25017686 DOI: 10.1016/j.iac.2014.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Several laboratories use the lymphocyte transformation test for the diagnosis of delayed-type drug hypersensitivity reactions. Recently, the availability of multiple readouts has improved our ability to diagnose reactions. It is important to note that most published studies characterizing the usefulness of diagnostic tests utilize blood samples from well-defined test and control patient groups. The purpose of this article is to briefly summarize the cellular and chemical basis of delayed-type drug hypersensitivity reactions and to review in vitro assays that are available for drug hypersensitivity diagnosis.
Collapse
Affiliation(s)
- Dean J Naisbitt
- Department of Clinical and Molecular Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England.
| | - Ryan G Nattrass
- Department of Clinical and Molecular Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England
| | - Monday O Ogese
- Department of Clinical and Molecular Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Sherrington Building, Ashton Street, Liverpool L69 3GE, England
| |
Collapse
|
18
|
The importance of hapten-protein complex formation in the development of drug allergy. Curr Opin Allergy Clin Immunol 2015; 14:293-300. [PMID: 24936850 DOI: 10.1097/aci.0000000000000078] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Drug allergy is an adverse drug reaction that is immune-mediated. Immune activation can occur when drugs or haptens bind covalently to proteins and then act as antigens. The purpose of this review is to summarize the recent data on the formation of hapten-protein complexes and to assess the importance of these complexes in the generation of drug allergy. RECENT FINDINGS The formation of hapten-protein complexes by drugs and their reactive metabolites has largely been investigated using model proteins such as human serum albumin. Precise identification of the structure of the hapten and the resulting modified residue(s) in the protein has been undertaken for a small number of drugs, such as p-phenylenediamine, nevirapine, carbamazepine, β-lactams and abacavir. Some progress has also been made in identifying hapten-protein complexes in the serum of patients with allergy. SUMMARY Drug-specific T cells have been isolated from different patients with allergy. Formation of hapten-protein complexes, their processing and antigen presentation have been implicated in the development of drug allergy to p-phenylenediamine, sulfonamides and β-lactams. However, evidence also supports the pi mechanism of immune activation wherein drugs interact directly with immune receptors. Thus, multiple mechanisms of immune activation may occur for the same drug.
Collapse
|
19
|
Roggen EL. In VitroApproaches for Detection of Chemical Sensitization. Basic Clin Pharmacol Toxicol 2014; 115:32-40. [DOI: 10.1111/bcpt.12202] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 01/14/2014] [Indexed: 11/30/2022]
|
20
|
van Haandel L, Goldman JL, Pearce RE, Leeder JS. Urinary Biomarkers of Trimethoprim Bioactivation in Vivo Following Therapeutic Dosing in Children. Chem Res Toxicol 2014; 27:211-8. [DOI: 10.1021/tx4003325] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Leon van Haandel
- Division of Clinical Pharmacology
and Therapeutic Innovation, Children’s
Mercy Hospitals and Clinics, and ‡Department of Pediatrics, School of Medicine, University of Missouri Kansas City, 2401 Gillham Road, Kansas
City, Missouri 64108, United States
| | - Jennifer. L. Goldman
- Division of Clinical Pharmacology
and Therapeutic Innovation, Children’s
Mercy Hospitals and Clinics, and ‡Department of Pediatrics, School of Medicine, University of Missouri Kansas City, 2401 Gillham Road, Kansas
City, Missouri 64108, United States
| | - Robin E. Pearce
- Division of Clinical Pharmacology
and Therapeutic Innovation, Children’s
Mercy Hospitals and Clinics, and ‡Department of Pediatrics, School of Medicine, University of Missouri Kansas City, 2401 Gillham Road, Kansas
City, Missouri 64108, United States
| | - J. Steven Leeder
- Division of Clinical Pharmacology
and Therapeutic Innovation, Children’s
Mercy Hospitals and Clinics, and ‡Department of Pediatrics, School of Medicine, University of Missouri Kansas City, 2401 Gillham Road, Kansas
City, Missouri 64108, United States
| |
Collapse
|
21
|
Uetrecht J, Naisbitt DJ. Idiosyncratic adverse drug reactions: current concepts. Pharmacol Rev 2013; 65:779-808. [PMID: 23476052 DOI: 10.1124/pr.113.007450] [Citation(s) in RCA: 199] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Idiosyncratic drug reactions are a significant cause of morbidity and mortality for patients; they also markedly increase the uncertainty of drug development. The major targets are skin, liver, and bone marrow. Clinical characteristics suggest that IDRs are immune mediated, and there is substantive evidence that most, but not all, IDRs are caused by chemically reactive species. However, rigorous mechanistic studies are very difficult to perform, especially in the absence of valid animal models. Models to explain how drugs or reactive metabolites interact with the MHC/T-cell receptor complex include the hapten and P-I models, and most recently it was found that abacavir can interact reversibly with MHC to alter the endogenous peptides that are presented to T cells. The discovery of HLA molecules as important risk factors for some IDRs has also significantly contributed to our understanding of these adverse reactions, but it is not yet clear what fraction of IDRs have a strong HLA dependence. In addition, with the exception of abacavir, most patients who have the HLA that confers a higher IDR risk with a specific drug will not have an IDR when treated with that drug. Interindividual differences in T-cell receptors and other factors also presumably play a role in determining which patients will have an IDR. The immune response represents a delicate balance, and immune tolerance may be the dominant response to a drug that can cause IDRs.
Collapse
Affiliation(s)
- Jack Uetrecht
- Faculties of Pharmacy and Medicine, University of Toronto, Toronto, Canada M5S3M2.
| | | |
Collapse
|
22
|
Evaluation of polymorphisms in the sulfonamide detoxification genes NAT2, CYB5A, and CYB5R3 in patients with sulfonamide hypersensitivity. Pharmacogenet Genomics 2013; 22:733-40. [PMID: 22850190 DOI: 10.1097/fpc.0b013e328357a735] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To determine whether polymorphisms in the sulfonamide detoxification genes, CYB5A (encoding cytochrome b(5)), CYB5R3 (encoding cytochrome b(5) reductase), or NAT2 (encoding N-acetyltransferase 2) were over-represented in patients with delayed sulfonamide drug hypersensitivity, compared with control patients who tolerated a therapeutic course of trimethoprim-sulfamethoxazole without adverse event. METHODS DNA from 99 nonimmunocompromised patients with sulfonamide hypersensitivity who were identified from the Personalized Medicine Research Project at the Marshfield Clinic, and from 99 age-matched, race-matched, and sex-matched drug-tolerant controls, were genotyped for four CYB5A and five CYB5R3 polymorphisms, and for all coding NAT2 SNPs. RESULTS CYB5A and CYB5R3 SNPs were found at low allele frequencies (<3-4%), which did not differ between hypersensitive and tolerant patients. NAT2 allele and haplotype frequencies, as well as inferred NAT2 phenotypes, also did not differ between groups (60 vs. 59% slow acetylators). Finally, no difference in NAT2 status was found in a subset of patients with more severe hypersensitivity signs (drug reaction with eosinophilia and systemic symptoms) compared with tolerant patients. CONCLUSION We found no evidence of a substantial involvement of these nine CYB5A or CYB5R3 polymorphisms in sulfonamide hypersensitivity risk, although minor effects cannot be completely ruled out. Despite careful medical record review and full resequencing of the NAT2 coding region, we found no association of NAT2 coding alleles with sulfonamide hypersensitivity (predominantly cutaneous eruptions) in this adult Caucasian population.
Collapse
|
23
|
Stachulski AV, Baillie TA, Kevin Park B, Scott Obach R, Dalvie DK, Williams DP, Srivastava A, Regan SL, Antoine DJ, Goldring CEP, Chia AJL, Kitteringham NR, Randle LE, Callan H, Castrejon JL, Farrell J, Naisbitt DJ, Lennard MS. The Generation, Detection, and Effects of Reactive Drug Metabolites. Med Res Rev 2012; 33:985-1080. [DOI: 10.1002/med.21273] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Andrew V. Stachulski
- Department of Chemistry, Robert Robinson Laboratories; University of Liverpool; Liverpool; L69 7ZD; UK
| | - Thomas A. Baillie
- School of Pharmacy; University of Washington; Box 357631; Seattle; Washington; 98195-7631
| | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - R. Scott Obach
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; Groton; Connecticut 06340
| | - Deepak K. Dalvie
- Pharmacokinetics, Dynamics and Metabolism; Pfizer Worldwide Research & Development; La Jolla; California 94121
| | - Dominic P. Williams
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Abhishek Srivastava
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Sophie L. Regan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Daniel J. Antoine
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Christopher E. P. Goldring
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Alvin J. L. Chia
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Neil R. Kitteringham
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Laura E. Randle
- School of Pharmacy and Biomolecular Sciences, Faculty of Science; Liverpool John Moores University; James Parsons Building, Byrom Street; Liverpool L3 3AF; UK
| | - Hayley Callan
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - J. Luis Castrejon
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - John Farrell
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology; MRC Centre for Drug Safety Science; Institute of Translational Medicine; University of Liverpool; Sherrington Buildings, Ashton Street; Liverpool L69 3GE; UK
| | - Martin S. Lennard
- Academic Unit of Medical Education; University of Sheffield; 85 Wilkinson Street; Sheffield S10 2GJ; UK
| |
Collapse
|
24
|
Abstract
Adverse drug reactions (ADRs) complicate at least 5% of all courses of therapy for children. Dealing with an ADR requires a stepwise approach in appreciation of the possibility of an ADR, assessment of whether the adverse event in question is drug-related, assessment of causality, assistance in treating the symptoms of the ADR, and dealing with the aftermath of the event. Several new developments likely will improve the ability to assess, evaluate, treat, and prevent ADRs in children. These developments include tools to evaluate causality, laboratory tests to diagnose ADRs, pharmacogenomic approaches to prevent ADRs, and new insights into treating serious ADRs.
Collapse
Affiliation(s)
- Michael Rieder
- Department of Paediatrics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
25
|
Faulkner L, Martinsson K, Santoyo-Castelazo A, Cederbrant K, Schuppe-Koistinen I, Powell H, Tugwood J, Naisbitt DJ, Park BK. The development of in vitro culture methods to characterize primary T-cell responses to drugs. Toxicol Sci 2012; 127:150-8. [PMID: 22331489 DOI: 10.1093/toxsci/kfs080] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Adverse drug reactions represent a major stumbling block to drug development and those with an immune etiology are the most difficult to predict. We have developed an in vitro T-cell priming culture method using peripheral blood from healthy volunteers to assess the allergenic potential of drugs. The drug metabolite nitroso sulfamethoxazole (SMX-NO) was used as a model drug allergen to establish optimum assay conditions. Naive T cells were cocultured with monocyte-derived dendritic cells at a ratio of 25:1 in the presence of the drug for a period of 8 days, to expand the number of drug-responsive T cells. The T cells were then incubated with fresh dendritic cells, and drug and their antigen responsiveness analyzed using readouts for proliferation, cytokine secretion, and cell phenotype. All five volunteers showed dose-dependent proliferation as measured by 5-(and 6)-carboxyfluorescein diacetate succinimidyl ester content and by (3)H-thymidine uptake. CD4 T cells that had divided in the presence of SMX-NO had changed from a naive phenotype (CD45RA+) to a memory phenotype (CD45RO+). These memory T cells expressed the chemokine receptors CCR2, CCR4, and CXCR3 suggesting a mixture of T(H)1 and T(H)2 cells in the responding population, with a propensity for homing to the skin. Drug stimulation was also associated with the secretion of a mixture of T(H)1 cytokines (interferon γ) and T(H)2 cytokines (interleukin [IL]-5 and IL-13) as detected by ELISpot. We are currently developing this approach to investigate the allergenic potential of other drugs, including those where an association between specific human leucocyte antigen alleles and susceptibility to an immunological reaction has been established.
Collapse
Affiliation(s)
- Lee Faulkner
- Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Eyanagi R, Toda A, Imoto M, Uchiyama H, Ishii Y, Kuroki H, Kuramoto Y, Soeda S, Shimeno H. Covalent binding of nitroso-sulfonamides to glutathione S-transferase in guinea pigs with delayed type hypersensitivity. Int Immunopharmacol 2012; 12:694-700. [PMID: 22342371 DOI: 10.1016/j.intimp.2012.01.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 01/20/2012] [Accepted: 01/31/2012] [Indexed: 01/22/2023]
Abstract
Drug induced allergies are believed to be induced by conjugates consisting of biological macromolecules and active metabolites. The present study investigated whether guinea pig glutathione S-transferase (gpGST), a protein that binds with sulfanilamide (SA) and sulfamethoxazole (SMX), could be detected in the liver cytosol fraction of guinea pigs that intraperitoneally received SA or SMX, and whether gpGST is a carrier protein. We synthesized three nitroso compounds, i.e., 4-nitroso-sulfanilamide (SA-NO), 4-nitrososulfamethoxazole (SMX-NO) and fluorescent-labeled nitroso compound (DNSBA-NO), and examined binding quantities of nitroso compounds to gpGST purified from untreated female guinea pigs. Furthermore, the concentrations of IgG in serum antibody for nitroso compounds were estimated using ELISA. When guinea pigs were sensitized using the three nitroso compounds, the dose dependent skin reactions were confirmed with each compound. In addition, sensitized guinea pigs using each nitroso compound showed positive skin reactions at an elicitation test performed using gpGST alone. The results confirmed synthesis of antibody against gpGST due to hapten sensitization. Therefore, when a nitroso compound binds with gpGST in the body of guinea pigs, nitroso-gpGST acts as a neoantigen, which induces synthesis of autoantibody. Thus, gpGST appears to be one of the carrier proteins that induce sulfa drug-induced allergies. Immunization of guinea pigs with active metabolite of drugs may give information for predicting the occurrence of delayed type hypersensitivity in human.
Collapse
Affiliation(s)
- Reiko Eyanagi
- Daiichi College of Pharmaceutical Sciences, 22-1 Tamagawa-cho, minami-ku, Fukuoka 815-8511, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Elsheikh A, Castrejon L, Lavergne SN, Whitaker P, Monshi M, Callan H, El-Ghaiesh S, Farrell J, Pichler WJ, Peckham D, Park BK, Naisbitt DJ. Enhanced antigenicity leads to altered immunogenicity in sulfamethoxazole-hypersensitive patients with cystic fibrosis. J Allergy Clin Immunol 2011; 127:1543-51.e3. [PMID: 21354601 DOI: 10.1016/j.jaci.2010.12.1119] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 11/09/2010] [Accepted: 12/28/2010] [Indexed: 11/26/2022]
Abstract
BACKGROUND Exposure of patients with cystic fibrosis to sulfonamides is associated with a high incidence of hypersensitivity reactions. OBJECTIVE To compare mechanisms of antigen presentation and characterize the phenotype and function of T cells from sulfamethoxazole-hypersensitive patients with and without cystic fibrosis. METHODS T cells were cloned from 6 patients and characterized in terms of phenotype and function. Antigen specificity and mechanisms of antigen presentation to specific clones were then explored. Antigen-presenting cell metabolism of sulfamethoxazole was quantified by ELISA. The involvement of metabolism in antigen presentation was evaluated by using enzyme inhibitors. RESULTS Enzyme inhibitable sulfamethoxazole-derived protein adducts were detected in antigen-presenting cells from patients with and without cystic fibrosis. A significantly higher quantity of adducts were detected with cells from patients with cystic fibrosis. Over 500 CD4(+) or CD8(+) T-cell clones were generated and shown to proliferate and kill target cells. Three patterns of MHC-restricted reactivity (sulfamethoxazole-responsive, sulfamethoxazole metabolite-responsive, and cross-reactive) were observed with clones from patients without cystic fibrosis. From patients with cystic fibrosis, sulfamethoxazole metabolite-responsive and cross-reactive, but not sulfamethoxazole-responsive, clones were observed. The response of the cross-reactive clones to sulfamethoxazole was dependent on adduct formation and was blocked by glutathione and enzyme inhibitors. Antigen-stimulated clones from patients with cystic fibrosis secreted higher levels of IFN-γ, IL-6, and IL-10, but lower levels of IL-17. CONCLUSION Sulfamethoxazole metabolism and protein adduct formation is critical for the stimulation of T cells from patients with cystic fibrosis. T cells from patients with cystic fibrosis secrete high levels of IFN-γ, IL-6, and IL-10.
Collapse
Affiliation(s)
- Ayman Elsheikh
- MRC Centre for Drug Safety Science, Department of Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Elsheikh A, Lavergne SN, Castrejon JL, Farrell J, Wang H, Sathish J, Pichler WJ, Park BK, Naisbitt DJ. Drug antigenicity, immunogenicity, and costimulatory signaling: evidence for formation of a functional antigen through immune cell metabolism. THE JOURNAL OF IMMUNOLOGY 2010; 185:6448-60. [PMID: 20980635 DOI: 10.4049/jimmunol.1000889] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recognition of drugs by immune cells is usually explained by the hapten model, which states that endogenous metabolites bind irreversibly to protein to stimulate immune cells. Synthetic metabolites interact directly with protein-generating antigenic determinants for T cells; however, experimental evidence relating intracellular metabolism in immune cells and the generation of physiologically relevant Ags to functional immune responses is lacking. The aim of this study was to develop an integrated approach using animal and human experimental systems to characterize sulfamethoxazole (SMX) metabolism-derived antigenic protein adduct formation in immune cells and define the relationship among adduct formation, cell death, costimulatory signaling, and stimulation of a T cell response. Formation of SMX-derived adducts in APCs was dose and time dependent, detectable at nontoxic concentrations, and dependent on drug-metabolizing enzyme activity. Adduct formation above a threshold induced necrotic cell death, dendritic cell costimulatory molecule expression, and cytokine secretion. APCs cultured with SMX for 16 h, the time needed for drug metabolism, stimulated T cells from sensitized mice and lymphocytes and T cell clones from allergic patients. Enzyme inhibition decreased SMX-derived protein adduct formation and the T cell response. Dendritic cells cultured with SMX and adoptively transferred to recipient mice initiated an immune response; however, T cells were stimulated with adducts derived from SMX metabolism in APCs, not the parent drug. This study shows that APCs metabolize SMX; subsequent protein binding generates a functional T cell Ag. Adduct formation above a threshold stimulates cell death, which provides a maturation signal for dendritic cells.
Collapse
Affiliation(s)
- Ayman Elsheikh
- Department of Pharmacology, Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Castrejon JL, Berry N, El-Ghaiesh S, Gerber B, Pichler WJ, Park BK, Naisbitt DJ. Stimulation of human T cells with sulfonamides and sulfonamide metabolites. J Allergy Clin Immunol 2010; 125:411-418.e4. [PMID: 20159253 DOI: 10.1016/j.jaci.2009.10.031] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 10/16/2009] [Accepted: 10/19/2009] [Indexed: 10/19/2022]
Abstract
BACKGROUND Exposure to sulfonamides is associated with a high incidence of hypersensitivity reactions. Antigen-specific T cells are involved in the pathogenesis; however, the nature of the antigen interacting with specific T-cell receptors is not fully defined. OBJECTIVE We sought to explore the frequency of sulfamethoxazole (SMX)- and SMX metabolite-specific T cells in hypersensitive patients, delineate the specificity of clones, define mechanisms of presentation, and explore additional reactivity with structurally related sulfonamide metabolites. METHODS SMX- and SMX metabolite-specific T-cell clones were generated from 3 patients. Antigen specificity, mechanisms of antigen presentation, and cross-reactivity of specific clones were then explored. Low-lying energy conformations of drugs (metabolites) were modeled, and the energies available for protein binding was estimated. RESULTS Lymphocytes proliferated with parent drugs (SMX, sulfadiazine, and sulfapyridine) and both hydroxylamine and nitroso metabolites. Three patterns of drug (metabolite) stimulation were seen: 44% were SMX metabolite specific, 43% were stimulated with SMX metabolites and SMX, and 14% were stimulated with SMX alone. Most metabolite-responsive T cells were stimulated with nitroso SMX-modified protein through a hapten mechanism involving processing. In contrast to SMX-responsive clones, which were highly specific, greater than 50% of nitroso SMX-specific clones were stimulated with nitroso metabolites of sulfapyridine and sulfadiazine but not nitrosobenzene. Pharmacophore modeling showed that the summation of available binding energies for protein interactions and the preferred spatial arrangement of atoms in each molecule determine a drug's potential to stimulate specific T cells. CONCLUSIONS Nitroso sulfonamide metabolites form potent antigenic determinants for T cells from hypersensitive patients. T-cell responses against drugs (metabolites) bound directly to MHC or MHC/peptide complexes can occur through cross-reactivity with the haptenic immunogen.
Collapse
Affiliation(s)
- J Luis Castrejon
- MRC Centre for Drug Safety Science, Department of Pharmacology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
30
|
Bhusari S, Abouraya M, Padilla ML, Pinkerton ME, Drescher NJ, Sacco JC, Trepanier LA. Combined ascorbate and glutathione deficiency leads to decreased cytochrome b5 expression and impaired reduction of sulfamethoxazole hydroxylamine. Arch Toxicol 2010; 84:597-607. [PMID: 20221587 DOI: 10.1007/s00204-010-0530-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Accepted: 02/22/2010] [Indexed: 10/19/2022]
Abstract
Sulfonamide antimicrobials such as sulfamethoxazole (SMX) have been associated with drug hypersensitivity reactions, particularly in patients with AIDS. A reactive oxidative metabolite, sulfamethoxazole-nitroso (SMX-NO), forms drug-tissue adducts that elicit a T-cell response. Antioxidants such as ascorbic acid (AA) and glutathione (GSH) reduce SMX-NO to the less reactive hydroxylamine metabolite (SMX-HA), which is further reduced to the non-immunogenic parent compound by cytochrome b (5) (b5) and its reductase (b5R). We hypothesized that deficiencies in AA and GSH would enhance drug-tissue adduct formation and immunogenicity toward SMX-NO and that these antioxidant deficiencies might also impair the activity of the b5/b5R pathway. We tested these hypotheses in guinea pigs fed either a normal or AA-restricted diet, followed by buthionine sulfoximine treatment (250 mg/kg SC daily, or vehicle); and SMX-NO (1 mg/kg IP 4 days per week, or vehicle), for 2 weeks. Guinea pigs did not show any biochemical or histopathologic evidence of SMX-NO-related toxicity. Combined AA and GSH deficiency in this model did not significantly increase tissue-drug adduct formation, or splenocyte proliferation in response to SMX-NO. However, combined antioxidant deficiency was associated with decreased mRNA and protein expression of cytochrome b (5), as well as significant decreases in SMX-HA reduction in SMX-NO-treated pigs. These results suggest that SMX-HA detoxification may be down-regulated in combined AA and GSH deficiency. This mechanism could contribute to the higher risk of SMX hypersensitivity in patients with AIDS with antioxidant depletion.
Collapse
Affiliation(s)
- Sachin Bhusari
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706-1102, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Castrejon JL, Lavergne SN, El-Sheikh A, Farrell J, Maggs JL, Sabbani S, O’Neill PM, Park BK, Naisbitt DJ. Metabolic and Chemical Origins of Cross-Reactive Immunological Reactions to Arylamine Benzenesulfonamides: T-Cell Responses to Hydroxylamine and Nitroso Derivatives. Chem Res Toxicol 2009; 23:184-92. [DOI: 10.1021/tx900329b] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- J. Luis Castrejon
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool L69 3GE, United Kingdom, and Department of Chemistry, The University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - Sidonie N. Lavergne
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool L69 3GE, United Kingdom, and Department of Chemistry, The University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - Ayman El-Sheikh
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool L69 3GE, United Kingdom, and Department of Chemistry, The University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - John Farrell
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool L69 3GE, United Kingdom, and Department of Chemistry, The University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - James L. Maggs
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool L69 3GE, United Kingdom, and Department of Chemistry, The University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - Sunil Sabbani
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool L69 3GE, United Kingdom, and Department of Chemistry, The University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - Paul M. O’Neill
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool L69 3GE, United Kingdom, and Department of Chemistry, The University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - B. Kevin Park
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool L69 3GE, United Kingdom, and Department of Chemistry, The University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - Dean J. Naisbitt
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool L69 3GE, United Kingdom, and Department of Chemistry, The University of Liverpool, Liverpool L69 7ZD, United Kingdom
| |
Collapse
|
32
|
Callan HE, Jenkins RE, Maggs JL, Lavergne SN, Clarke SE, Naisbitt DJ, Park BK. Multiple adduction reactions of nitroso sulfamethoxazole with cysteinyl residues of peptides and proteins: implications for hapten formation. Chem Res Toxicol 2009; 22:937-48. [PMID: 19358516 DOI: 10.1021/tx900034r] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Sulfamethoxazole (SMX) induces immunoallergic reactions that are thought to be a result of intracellular protein haptenation by its nitroso metabolite (SMX-NO mass, 267 amu). SMX-NO reacts with protein thiols in vitro, but the conjugates have not been defined chemically. The reactions of SMX-NO with glutathione (GSH), a synthetic peptide (DS3), and two model proteins, human GSH S-transferase pi (GSTP) and serum albumin (HSA), were investigated by mass spectrometry. SMX-NO formed a semimercaptal (N-hydroxysulfenamide) conjugate with GSH that rearranged rapidly (1-5 min) to a sulfinamide. Reaction of SMX-NO with DS3 also yielded a sulfinamide adduct (mass increment, 267 amu) on the cysteine residue. GSTP was exclusively modified at the reactive Cys47 by SMX-NO and exhibited mass increments of 267, 283, and 299 amu, indicative of sulfinamide, N-hydroxysulfinamide, and N-hydroxysulfonamide adducts, respectively. HSA was modified at Cys34, forming only the N-hydroxysulfinamide adduct. HSA modification by SMX-NO under these conditions was confirmed with ELISA and immunoblotting with an antisulfonamide antibody. It is proposed that cysteine-linked N-hydroxysulfinamide and N-hydroxysulfonamide adducts of SMX are formed via the reaction of SMX-NO with cysteinyl sulfoxy acids. Evidence for a multistep assembly of model sulfonamide epitopes on GSH and polypeptides via hydrolyzable intermediates is also presented. In summary, novel, complex, and metastable haptenic structures have been identified on proteins exposed in vitro to the nitroso metabolite of SMX.
Collapse
Affiliation(s)
- Hayley E Callan
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, School of Biomedical Sciences, The University of Liverpool, Liverpool L69 3GE, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
33
|
[HLA-B*5701 and hypersensitivity reactions to abacavir. Study methods and clinical relevance]. Enferm Infecc Microbiol Clin 2008; 26 Suppl 6:34-9. [PMID: 18680694 DOI: 10.1016/s0213-005x(08)76510-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hypersensitivity reactions to abacavir occur in 5-8% of patients starting treatment with this drug and limits future treatment. Some host genetic factors, especially the HLA-B*5701 allele, have been identified as risk factors for hypersensitivity reaction in Caucasians. Consequently, the possibility of routine implementation of a genetic test to rule out the presence of this allele has been proposed to achieve a personalized therapeutic profile. The present article discusses all the information related to hypersensitivity to abacavir and its genetic and immunological markers, as well as the distinct techniques for HLA-B*5701 allele detection. The various studies performed to date in distinct population are also discussed.
Collapse
|
34
|
Cribb AE, Peyrou M, Muruganandan S, Schneider L. The Endoplasmic Reticulum in Xenobiotic Toxicity. Drug Metab Rev 2008; 37:405-42. [PMID: 16257829 DOI: 10.1080/03602530500205135] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The endoplasmic reticulum (ER) is involved in an array of cellular functions that play important roles in xenobiotic toxicity. The ER contains the majority of cytochrome P450 enzymes involved in xenobiotic metabolism, as well as a number of conjugating enzymes. In addition to its role in drug bioactivation and detoxification, the ER can be a target for damage by reactive intermediates leading to cell death or immune-mediated toxicity. The ER contains a set of luminal proteins referred to as ER stress proteins (including GRP78, GRP94, protein disulfide isomerase, and calreticulin). These proteins help regulate protein processing and folding of membrane and secretory proteins in the ER, calcium homeostasis, and ER-associated apoptotic pathways. They are induced in response to ER stress. This review discusses the importance of the ER in molecular events leading to cell death following xenobiotic exposure. Data showing that the ER is important in both renal and hepatic toxicity will be discussed.
Collapse
Affiliation(s)
- Alastair E Cribb
- Laboratory of Comparative Pharmacogenetics, Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PEI, Canada.
| | | | | | | |
Collapse
|
35
|
Eyanagi R, Toda A, Ishii Y, Saito H, Soeda S, Shimeno H, Shigematsu H. Antigenicity of sulfanilamide and its metabolites using fluorescent-labelled compounds. Xenobiotica 2008; 35:911-25. [PMID: 16308284 DOI: 10.1080/00498250500251533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In order to clarify the onset mechanisms of drug-induced allergies, three fluorescent-labelled compounds were synthesized by subjecting sulfanilamide (SA), a base compound for sulfonamides, and its active metabolites, i.e. sulfanilamide hydroxylamine and sulfanilamide nitroso, to dansylation using dansylchloride. In other words, 5-dimethylamino-N-(4-aminobenzyl)-naphthalenesulfonamide (DNS-4ABA), 5-dimethylamino-N-(4-hydroxylaminobenzyl)-1-naphthalenesulfonamide (DNS-4HABA) and 5-dimethylamino-N-(4-nitrosobenzyl)-1-naphthalenesulfonamide (DNS-4NSBA) were synthesized as model haptens. When analysed by HPLC, a conjugate of DNS-4HABA and glutathione (GSH) with nucleophilic amino acids had two peaks (P-1 and P-2). FAB-MS and 1H-NMR revealed that the DNS-4HABA-GSH conjugate consisted of sulphinamide and semimercaptal. The reactivity of GSH to DNS-4ABA, DNS-4HABA and DNS-4NSBA was quantified by HPLC using an oxidization system (horseradish peroxidase/H2O2). The results show that production of DNS-4NSBA-GSH-conjugate was four to eight times higher than that of DNS-4HABA-GSH conjugate, but that DNS-4ABA did not bind with GSH. Skin reactions were assessed using guinea pigs, and strong delayed erythema was seen with DNS-4NSBA, which bound most strongly with GSH, whereas weak delayed erythema was seen with DNS-4ABA, which did not bind with GSH. This suggests a correlation between GSH conjugate production and skin reactions. DNS-4HABA enzymatically bound with proteins in rat and guinea pig liver cytosol and microsomal fractions. The proteins that bound to DNS-4HABA were purified by HPLC and then subjected to N-terminal amino acid analysis. Ubiquitin (10 kDa) and fatty acid binding protein (30 kDa) were detected in the rat liver cytosol fraction; retinol-dehydrogenase (35 kDa) in the rat microsomal fraction; and glutathione-S-transferase B (mmu) (25 kDa) in the guinea pig liver cytosol fraction. When DNS-4HABA or DNS-4NSBA binds to proteins that play important roles in the body, unexpected adverse reactions may occur. Furthermore, by utilizing our technique using model compounds, it may be possible to identify the carrier proteins of various compounds, including pharmaceutical agents.
Collapse
Affiliation(s)
- R Eyanagi
- Daiichi College of Pharmaceutical Sciences, Fukuoka, Japan.
| | | | | | | | | | | | | |
Collapse
|
36
|
Cheng L, Stewart BJ, You Q, Petersen DR, Ware JA, Piccotti JR, Kawabata TT, Ju C. Covalent Binding of the Nitroso Metabolite of Sulfamethoxazole Is Important in Induction of Drug-Specific T-Cell Responses in Vivo. Mol Pharmacol 2008; 73:1769-75. [DOI: 10.1124/mol.107.043273] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
37
|
Almeida CAM, Martin AM, Nolan D, Lucas A, Cameron PU, James I, Phillips E, Mallal S. Cytokine Profiling in Abacavir Hypersensitivity Patients. Antivir Ther 2008. [DOI: 10.1177/135965350801300202] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background Abacavir hypersensitivity in genetically susceptible individuals implicates an abacavir-specific T-cell response to either the parent drug or a metabolite generated in vivo. We have analysed the cytokine profile in antigen-presenting cells and the T-lymphocytes that are involved in the pathological immune response to abacavir. Methods In this study, we compared abacavir-specific cytokine responses in cultured peripheral blood mononuclear cells (PBMCs) from HIV-infected abacavir hypersensitive, tolerant and naive individuals. Cells were cultured in the presence or absence of abacavir. Cytokine expression was determined by microarray analysis, enzyme-linked immunosorbent assays and flow cytometry. Results We demonstrated using in vitro models of immune activation that the production of interferon-γwas specifically induced by abacavir treatment in PBMCs obtained from hypersensitive patients carrying the HLA-B*5701 allele (median 123.86 compared with -30.83 for tolerant controls, P=0.001). Conclusion These results provide further insight into the immunological and metabolic basis of abacavir hypersensitivity syndrome. In vitro assays could assist in the identification of susceptible loci by providing a surrogate marker for the hypersensitivity reaction. Such a marker could be studied in unexposed individuals to shed further light on the immunopathogenesis of the abacavir hypersensitivity syndrome.
Collapse
Affiliation(s)
- Coral-Ann M Almeida
- Centre for Clinical Immunology and Biomedical Statistics, Royal Perth Hospital and Murdoch University, Perth, Australia
| | - Annalise M Martin
- Centre for Clinical Immunology and Biomedical Statistics, Royal Perth Hospital and Murdoch University, Perth, Australia
| | - David Nolan
- Centre for Clinical Immunology and Biomedical Statistics, Royal Perth Hospital and Murdoch University, Perth, Australia
| | - Andrew Lucas
- Centre for Clinical Immunology and Biomedical Statistics, Royal Perth Hospital and Murdoch University, Perth, Australia
| | - Paul U Cameron
- Department of Clinical Immunology and Biochemical Genetics, Royal Perth Hospital, Perth, Australia
| | - Ian James
- Centre for Clinical Immunology and Biomedical Statistics, Royal Perth Hospital and Murdoch University, Perth, Australia
| | - Elizabeth Phillips
- Centre for Clinical Immunology and Biomedical Statistics, Royal Perth Hospital and Murdoch University, Perth, Australia
| | - Simon Mallal
- Centre for Clinical Immunology and Biomedical Statistics, Royal Perth Hospital and Murdoch University, Perth, Australia
- Department of Clinical Immunology and Biochemical Genetics, Royal Perth Hospital, Perth, Australia
| |
Collapse
|
38
|
Sanderson JP, Naisbitt DJ, Farrell J, Ashby CA, Tucker MJ, Rieder MJ, Pirmohamed M, Clarke SE, Park BK. Sulfamethoxazole and its metabolite nitroso sulfamethoxazole stimulate dendritic cell costimulatory signaling. THE JOURNAL OF IMMUNOLOGY 2007; 178:5533-42. [PMID: 17442935 DOI: 10.4049/jimmunol.178.9.5533] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Different signals in addition to the antigenic signal are required to initiate an immunological reaction. In the context of sulfamethoxazole allergy, the Ag is thought to be derived from its toxic nitroso metabolite, but little is known about the costimulatory signals, including those associated with dendritic cell maturation. In this study, we demonstrate increased CD40 expression, but not CD80, CD83, or CD86, with dendritic cell surfaces exposed to sulfamethoxazole (250-500 microM) and the protein-reactive metabolite nitroso sulfamethoxazole (1-10 microM). Increased CD40 expression was not associated with apoptosis or necrosis, or glutathione depletion. Covalently modified intracellular proteins were detected when sulfamethoxazole was incubated with dendritic cells. Importantly, the enzyme inhibitor 1-aminobenzotriazole prevented the increase in CD40 expression with sulfamethoxazole, but not with nitroso sulfamethoxazole or LPS. The enzymes CYP2C9, CYP2C8, and myeloperoxidase catalyzed the conversion of sulfamethoxazole to sulfamethoxazole hydroxylamine. Myeloperoxidase was expressed at high levels in dendritic cells. Nitroso sulfamethoxazole immunogenicity was inhibited in mice with a blocking anti-CD40L Ab. In addition, when a primary nitroso sulfamethoxazole-specific T cell response using drug-naive human cells was generated, the magnitude of the response was enhanced when cultures were exposed to a stimulatory anti-CD40 Ab. Finally, increased CD40 expression was 5-fold higher on nitroso sulfamethoxazole-treated dendritic cells from an HIV-positive allergic patient compared with volunteers. These data provide evidence of a link between localized metabolism, dendritic cell activation, and drug immunogenicity.
Collapse
Affiliation(s)
- Joseph P Sanderson
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Naisbitt DJ, Pirmohamed M, Park BK. Immunological principles of T-cell-mediated adverse drug reactions in skin. Expert Opin Drug Saf 2007; 6:109-24. [PMID: 17367257 DOI: 10.1517/14740338.6.2.109] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Drug hypersensitivity reactions in skin are an immune-mediated phenomenon associated with significant patient mortality and morbidity. Antigen-specific T cells, which have been isolated from the peripheral circulation and target organs of hypersensitive patients, are thought to propagate and regulate the development of clinical symptoms. The investigation of clinical cases with respect to the basic cellular and chemical mechanisms that underpin drug hypersensitivity has resulted in: i) the need to redress some aspects of present immunological dogma; and ii) additional fundamental immunological questions. Thus, the aim of this review article is to summarise present opinion on how and why drugs initiate a pathogenic T-cell response in a small section of the population and subsequently reflect on gaps in basic immunology and where future research might lead.
Collapse
Affiliation(s)
- Dean J Naisbitt
- University of Liverpool, Department of Pharmacology, The Sherrington Building, Ashton Street, Liverpool, UK.
| | | | | |
Collapse
|
40
|
Roychowdhury S, Vyas PM, Svensson CK. Formation and Uptake of Arylhydroxylamine-Haptenated Proteins in Human Dendritic Cells. Drug Metab Dispos 2007; 35:676-81. [PMID: 17220235 DOI: 10.1124/dmd.106.013680] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Bioactivation of sulfonamides and the subsequent formation of haptenated proteins is believed to be a critical step in the development of hypersensitivity reactions to these drugs. Numerous lines of evidence suggest that the presence of such adducts in dendritic cells (DCs) migrating to draining lymph nodes is essential for the development of cutaneous reactions to xenobiotics. Our objective was to determine the ability of human DCs to form drug-protein covalent adducts when exposed to sulfamethoxazole (SMX), dapsone (DDS), or their arylhydroxylamine metabolites [sulfamethoxazole hydroxylamine (S-NOH) and dapsone hydroxylamine (D-NOH)] and to take up preformed adduct. Naive and immature CD34+ KG-1 cells were incubated with SMX, DDS, or metabolites. Formation of haptenated proteins was probed using confocal microscopy and ELISA. Cells were also incubated with preformed adduct (drug-bovine serum albumin conjugate), and uptake was determined using confocal microscopy. Both naive and immature KG-1 cells were able to bioactivate DDS, forming drug-protein adducts, whereas cells showed very little protein haptenation when exposed to SMX. Exposure to S-NOH or D-NOH resulted in protein haptenation in both cell types. Both immature and naive KG-1 cells were able to take up preformed haptenated proteins. Thus, DCs may acquire haptenated proteins associated with drugs via intracellular bioactivation, uptake of reactive metabolites, or uptake of adduct formed and released by adjacent cells (e.g., keratinocytes).
Collapse
Affiliation(s)
- Sanjoy Roychowdhury
- Office of the Dean, College of Pharmacy, Nursing and Health Sciences, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 49707, USA
| | | | | |
Collapse
|
41
|
Lin D, Tucker MJ, Rieder MJ. Increased adverse drug reactions to antimicrobials and anticonvulsants in patients with HIV infection. Ann Pharmacother 2006; 40:1594-601. [PMID: 16912251 DOI: 10.1345/aph.1g525] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To review the incidence, signs, symptoms, and mechanisms of adverse drug reactions (ADRs) to sulfonamides, anticonvulsants, and antimycobacterial medications among people with HIV. DATA SOURCES Searches of MEDLINE/PubMed (1980-November 2005) and National Library of Medicine Meeting Abstracts (1989-November 2005), as well as hand searches of journals and abstracts, were conducted to identify primary literature. Reference lists were reviewed to identify additional relevant reports. STUDY SELECTION AND DATA EXTRACTION Relevant articles and abstracts, particularly of in vitro experiments and clinical studies, were compiled and reviewed. DATA SYNTHESIS ADRs, especially in HIV-infected patients, are a cause for concern. Sulfonamides, anticonvulsants, and antimycobacterial drugs are commonly used to prevent and treat complications of HIV, including seizures and opportunistic infections. Patients with HIV have a much greater rate of ADRs to these drug classes, including severe and life-threatening hypersensitivity reactions. Several mechanisms of these ADRs have been postulated. Sulfamethoxazole and anticonvulsant hypersensitivity may involve the increased formation and decreased detoxification of reactive metabolites. The mechanisms for the marked increase in hypersensitivity ADRs to antimycobacterial drugs may be related to an altered immune profile in patients infected with both tuberculosis and HIV. CONCLUSIONS ADRs to antimicrobial and anticonvulsant therapy cause markedly increased morbidity and mortality in HIV-positive patients. Further research involving the interaction between HIV and the increased ADRs to these drugs is required.
Collapse
Affiliation(s)
- Daren Lin
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada
| | | | | |
Collapse
|
42
|
Lavergne SN, Danhof RS, Volkman EM, Trepanier LA. Association of drug-serum protein adducts and anti-drug antibodies in dogs with sulphonamide hypersensitivity: A naturally occurring model of idiosyncratic drug toxicity. Clin Exp Allergy 2006; 36:907-15. [PMID: 16839406 DOI: 10.1111/j.1365-2222.2006.02506.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Sulphonamide antimicrobials, such as sulphamethoxazole (SMX), provide effective infection prophylaxis in immunocompromised patients, but can lead to drug hypersensitivity (HS) reactions. These reactions also occur in dogs, with a similar time course and clinical presentation as seen in humans. OBJECTIVES Drug-serum adducts and anti-drug antibodies have been identified in sulphonamide HS humans. The aim of this study was to determine whether similar markers were present in dogs with sulphonamide HS. METHODS Thirty-four privately owned sulphonamide HS dogs, 10 sulphonamide-'tolerant' dogs, 18 sulphonamide-naïve dogs, and four dogs experimentally dosed with SMX and the oxidative metabolite SMX-nitroso, were tested for drug-serum adducts by immunoblotting, and anti-drug antibodies by ELISA. RESULTS Sulphonamide-serum adducts were found in 10/20 HS dogs tested (50%), but in no tolerant dogs. Anti-sulphonamide IgG antibodies were detected in 17/34 HS dogs (50%), but in only one tolerant dog; antibody absorbance values were significantly higher in HS dogs. There was a significant association between the presence of sulphonamide-serum adducts and anti-sulphonamide antibodies (P = 0.009). Anti-drug antibodies were also found in dogs experimentally dosed with SMX-nitroso followed by SMX, but not in a dog dosed with drug vehicle, followed by SMX. CONCLUSION Similar humoral markers are present in dogs and humans with sulphonamide HS, supporting the use of dogs as a naturally occurring model for this syndrome in humans. These data suggest the potential use of drug-serum adducts and anti-drug antibodies as markers for sulphonamide HS. Preliminary data indicate that anti-sulphonamide antibodies may be triggered by the SMX-nitroso metabolite, not by the parent drug, in dogs.
Collapse
Affiliation(s)
- S N Lavergne
- Department of Medical Sciences, University of Wisconsin-Madison, Madison, WI 53706-1102, USA
| | | | | | | |
Collapse
|
43
|
Lavergne SN, Kurian JR, Bajad SU, Maki JE, Yoder AR, Guzinski MV, Graziano FM, Trepanier LA. Roles of endogenous ascorbate and glutathione in the cellular reduction and cytotoxicity of sulfamethoxazole-nitroso. Toxicology 2006; 222:25-36. [PMID: 16473451 DOI: 10.1016/j.tox.2006.01.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2005] [Revised: 12/21/2005] [Accepted: 01/12/2006] [Indexed: 10/25/2022]
Abstract
Sulfamethoxazole (SMX) is an effective drug for the management of opportunistic infections, but its use is limited by hypersensitivity reactions, particularly in HIV-infected patients. The oxidative metabolite SMX-nitroso (SMX-NO), is thought to be a proximate mediator of SMX hypersensitivity, and can be reduced in vitro by ascorbate or glutathione. Leukocytes from patients with SMX hypersensitivity show enhanced cytotoxicity from SMX metabolites in vitro; this finding has been attributed to a possible "detoxification defect" in some individuals. The purpose of this study was to determine whether variability in endogenous ascorbate or glutathione could be associated with individual differences in SMX-NO cytotoxicity. Thirty HIV-positive patients and 23 healthy control subjects were studied. Both antioxidants were significantly correlated with the reduction of SMX-NO to its hydroxylamine, SMX-HA, by mononuclear leukocytes, and both were linearly depleted during reduction. Controlled ascorbate supplementation in three healthy subjects increased leukocyte ascorbate with no change in glutathione, and significantly enhanced SMX-NO reduction. Ascorbate supplementation also decreased SMX-NO cytotoxicity compared to pre-supplementation values. Rapid reduction of SMX-NO to SMX-HA was associated with enhanced direct cytotoxicity from SMX-NO. When forward oxidation of SMX-HA back to SMX-NO was driven by the superoxide dismutase mimetic, Tempol, SMX-NO cytotoxicity was increased, without enhancement of adduct formation. This suggests that SMX-NO cytotoxicity may be mediated, at least in part, by redox cycling between SMX-HA and SMX-NO. Overall, these data indicate that endogenous ascorbate and glutathione are important for the intracellular reduction of SMX-NO, a proposed mediator of SMX hypersensitivity, and that redox cycling of SMX-HA to SMX-NO may contribute to the cytotoxicity of these metabolites in vitro.
Collapse
Affiliation(s)
- Sidonie N Lavergne
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706-1102, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Bhaiya P, Roychowdhury S, Vyas PM, Doll MA, Hein DW, Svensson CK. Bioactivation, protein haptenation, and toxicity of sulfamethoxazole and dapsone in normal human dermal fibroblasts. Toxicol Appl Pharmacol 2006; 215:158-67. [PMID: 16603214 PMCID: PMC1615915 DOI: 10.1016/j.taap.2006.02.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 02/02/2006] [Accepted: 02/02/2006] [Indexed: 01/19/2023]
Abstract
Cutaneous drug reactions (CDRs) associated with sulfonamides are believed to be mediated through the formation of reactive metabolites that result in cellular toxicity and protein haptenation. We evaluated the bioactivation and toxicity of sulfamethoxazole (SMX) and dapsone (DDS) in normal human dermal fibroblasts (NHDF). Incubation of cells with DDS or its metabolite (D-NOH) resulted in protein haptenation readily detected by confocal microscopy and ELISA. While the metabolite of SMX (S-NOH) haptenated intracellular proteins, adducts were not evident in incubations with SMX. Cells expressed abundant N-acetyltransferase-1 (NAT1) mRNA and activity, but little NAT2 mRNA or activity. Neither NAT1 nor NAT2 protein was detected. Incubation of NHDF with S-NOH or D-NOH increased reactive oxygen species formation and reduced glutathione content. NHDF were less susceptible to the cytotoxic effect of S-NOH and D-NOH than are keratinocytes. Our studies provide the novel observation that NHDF are able to acetylate both arylamine compounds and bioactivate the sulfone DDS, giving rise to haptenated proteins. The reactive metabolites of SMX and DDS also provoke oxidative stress in these cells in a time- and concentration-dependent fashion. Further work is needed to determine the role of the observed toxicity in mediating CDRs observed with these agents.
Collapse
Affiliation(s)
- Payal Bhaiya
- Division of Pharmaceutics, College of Pharmacy, The University of Iowa, Iowa City, IA
| | - Sanjoy Roychowdhury
- Division of Pharmaceutics, College of Pharmacy, The University of Iowa, Iowa City, IA
| | - Piyush M. Vyas
- Division of Pharmaceutics, College of Pharmacy, The University of Iowa, Iowa City, IA
| | - Mark A. Doll
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY
| | - David W. Hein
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY
| | - Craig K. Svensson
- Division of Pharmaceutics, College of Pharmacy, The University of Iowa, Iowa City, IA
| |
Collapse
|
45
|
Martin A, Nolan D, Almeida CA, Rauch A, Mallal S. Predicting and diagnosing abacavir and nevirapine drug hypersensitivity: from bedside to bench and back again. Pharmacogenomics 2006; 7:15-23. [PMID: 16354121 DOI: 10.2217/14622416.7.1.15] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
There is a growing discussion surrounding the issue of personalized approaches to drug prescription based on an individual's genetic makeup. This field of investigation has focused primarily on identifying genetic factors that influence drug metabolism and cellular disposition, thereby contributing to dose-dependent toxicities and/or variable drug efficacy. However, pharmacogenetic approaches have also proved valuable in predicting drug hypersensitivity reactions in selected patient populations, including HIV-infected patients receiving long-term antiretroviral therapy. In this instance, susceptibility has been strongly linked to genetic loci involved in antigen recognition and presentation to the immune system--most notably within the major histocompatibility complex (MHC) region--consistent with the notion that hypersensitivity reactions represent drug-specific immune responses that are largely dose independent. Here the authors describe their experiences with the development of pharmacogenetic approaches to hypersensitivity reactions associated with abacavir and nevirapine, two commonly prescribed antiretroviral drugs. It is demonstrated that prospective screening tests to identify and exclude individuals with a certain genetic makeup may be largely successful in decreasing or eliminating incidence of these adverse drug reactions in certain populations. This review also explores the broader implications of these findings.
Collapse
Affiliation(s)
- Annalise Martin
- Royal Perth Hospital and Murdoch University, Centre for Clinical Immunology and Biomedical Statistics, 2nd floor, North Block, Perth, 6000 Western Australia
| | | | | | | | | |
Collapse
|
46
|
Sanderson JP, Naisbitt DJ, Park BK. Role of bioactivation in drug-induced hypersensitivity reactions. AAPS JOURNAL 2006; 8:E55-64. [PMID: 16584134 PMCID: PMC2751424 DOI: 10.1208/aapsj080107] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Drug-induced hypersensitivity reactions are a major problem in both clinical treatment and drug development. This review covers recent developments in our understanding of the pathogenic mechanisms involved, with special focus on the potential role of metabolism and bioactivation in generating a chemical signal for activation of the immune system. The possible role of haptenation and neoantigen formation is discussed, alongside recent findings that challenge this paradigm. Additionally, the essential role of costimulation is examined, as are the potential points whereby costimulation may be driven by reactive metabolites. The relevance of local generation of metabolites in determining the location and character of a reaction is also covered.
Collapse
Affiliation(s)
- Joseph P. Sanderson
- Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, L69 3GE Liverpool, England
| | - Dean J. Naisbitt
- Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, L69 3GE Liverpool, England
| | - B. Kevin Park
- Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, L69 3GE Liverpool, England
| |
Collapse
|
47
|
Vyas PM, Roychowdhury S, Svensson CK. ROLE OF HUMAN CYCLOOXYGENASE-2 IN THE BIOACTIVATION OF DAPSONE AND SULFAMETHOXAZOLE. Drug Metab Dispos 2005; 34:16-8. [PMID: 16214851 DOI: 10.1124/dmd.105.006890] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sulfamethoxazole (SMX) and dapsone (4,4'-diaminodiphenylsulfone, DDS) are believed to mediate their adverse effects subsequent to bioactivation to their respective arylhydroxylamine and arylnitroso metabolites, resulting in covalent adduct formation with intracellular proteins. Various bioactivating enzymes, such as cytochromes P450 and myeloperoxidase, have been shown to be capable of catalyzing the N-oxidation of these compounds. We assessed the role of human cyclooxygenase-2 (COX-2) in the metabolism and subsequent adduct formation of DDS and SMX using recombinant human COX-2. Using an adduct-specific enzyme-linked immunosorbent assay, we found that the complete enzyme system gave rise to covalent adducts. However, the nonspecific COX inhibitor indomethacin did not reduce the amount of covalent adduct formed. Formation of the arylhydroxylamine metabolites was demonstrated via high performance liquid chromatography coupled with UV absorption. Metabolite formation was found to be secondary to the H2O2 in the incubation mixture and was not enzyme-mediated. Hence, COX-2 does not play a direct role in the bioactivation of these parent drugs to their arylhydroxylamine metabolites.
Collapse
Affiliation(s)
- Piyush M Vyas
- Division of Pharmaceutics, College of Pharmacy, The University of Iowa, 115 S. Grand Avenue, S213 PHAR, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
48
|
Zhou S, Chan E, Duan W, Huang M, Chen YZ. Drug bioactivation, covalent binding to target proteins and toxicity relevance. Drug Metab Rev 2005; 37:41-213. [PMID: 15747500 DOI: 10.1081/dmr-200028812] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A number of therapeutic drugs with different structures and mechanisms of action have been reported to undergo metabolic activation by Phase I or Phase II drug-metabolizing enzymes. The bioactivation gives rise to reactive metabolites/intermediates, which readily confer covalent binding to various target proteins by nucleophilic substitution and/or Schiff's base mechanism. These drugs include analgesics (e.g., acetaminophen), antibacterial agents (e.g., sulfonamides and macrolide antibiotics), anticancer drugs (e.g., irinotecan), antiepileptic drugs (e.g., carbamazepine), anti-HIV agents (e.g., ritonavir), antipsychotics (e.g., clozapine), cardiovascular drugs (e.g., procainamide and hydralazine), immunosupressants (e.g., cyclosporine A), inhalational anesthetics (e.g., halothane), nonsteroidal anti-inflammatory drugs (NSAIDSs) (e.g., diclofenac), and steroids and their receptor modulators (e.g., estrogens and tamoxifen). Some herbal and dietary constituents are also bioactivated to reactive metabolites capable of binding covalently and inactivating cytochrome P450s (CYPs). A number of important target proteins of drugs have been identified by mass spectrometric techniques and proteomic approaches. The covalent binding and formation of drug-protein adducts are generally considered to be related to drug toxicity, and selective protein covalent binding by drug metabolites may lead to selective organ toxicity. However, the mechanisms involved in the protein adduct-induced toxicity are largely undefined, although it has been suggested that drug-protein adducts may cause toxicity either through impairing physiological functions of the modified proteins or through immune-mediated mechanisms. In addition, mechanism-based inhibition of CYPs may result in toxic drug-drug interactions. The clinical consequences of drug bioactivation and covalent binding to proteins are unpredictable, depending on many factors that are associated with the administered drugs and patients. Further studies using proteomic and genomic approaches with high throughput capacity are needed to identify the protein targets of reactive drug metabolites, and to elucidate the structure-activity relationships of drug's covalent binding to proteins and their clinical outcomes.
Collapse
Affiliation(s)
- Shufeng Zhou
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.
| | | | | | | | | |
Collapse
|
49
|
Roychowdhury S, Vyas PM, Reilly TP, Gaspari AA, Svensson CK. Characterization of the Formation and Localization of Sulfamethoxazole and Dapsone-Associated Drug-Protein Adducts in Human Epidermal Keratinocytes. J Pharmacol Exp Ther 2005; 314:43-52. [PMID: 15784651 DOI: 10.1124/jpet.105.086009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sulfonamide- and sulfone-induced hypersensitivity reactions are thought to be mediated through bioactivation of parent drug molecule(s) to their respective reactive metabolite(s). Recent studies have demonstrated that keratinocytes can bioactivate sulfonamides and sulfones. Using enzyme-linked immunosorbent assay and hapten-specific rabbit antisera developed in our laboratory, we found that incubation of either normal human epidermal keratinocytes (NHEKs) or an immortalized human keratinocyte cell line (HaCaT) with sulfamethoxazole (SMX) or dapsone (DDS) resulted in the formation of drug/metabolite protein adducts. The formation of these adducts with SMX was increased in the presence of ascorbic acid, whereas N-acetylcysteine decreased adduct formation with both SMX and DDS. Adduct formation was confirmed using confocal microscopy when NHEKs were incubated with SMX, DDS, or their respective arylhydroxylamine metabolites. Cellular distribution of adducts was compared in permeable versus nonpermeable NHEKs. Exposure to SMX, DDS, or dapsone hydroxylamine resulted in the formation of intracellular adducts, whereas SMX hydroxylamine also resulted in the presence of adducts on the cell surface. In summary, our work shows that keratinocytes can bioactivate SMX/DDS to form drug-protein adducts, which may be acquired by antigen-presenting cells upon keratinocyte cell death, evoking an immune response. In addition, keratinocytes may themselves present antigen to hapten-specific cytotoxic T lymphocytes. Furthermore, our results also suggest that different sulfonamides/sulfones may have different protein targets for in situ haptenation in keratinocytes.
Collapse
Affiliation(s)
- Sanjoy Roychowdhury
- Division of Pharmaceutics, College of Pharmacy, The University of Iowa, 115 South Grand Avenue, S213 PHAR, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
50
|
Lavergne SN, Volkman EM, Maki JE, Yoder AR, Trepanier LA. Evaluation of the clinical, immunologic, and biochemical effects of nitroso sulfamethoxazole administration to dogs: a pilot study. Toxicology 2005; 208:63-72. [PMID: 15664433 DOI: 10.1016/j.tox.2004.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2004] [Revised: 11/04/2004] [Accepted: 11/04/2004] [Indexed: 11/21/2022]
Abstract
Sulfonamide antimicrobials such as sulfamethoxazole (SMX) have been associated in humans with hypersensitivity reactions, to include fever, skin eruptions, hepatotoxicity, and blood dyscrasias. These reactions also occur in dogs, the only non-human species known to develop a similar spectrum of sulfonamide hypersensitivity. Sulfonamide hypersensitivity is not well understood, but has been hypothesized to be due to the generation of the reactive oxidative metabolite, nitroso sulfamethoxazole (SMX-NO). SMX-NO, unlike the parent sulfonamide, is cytotoxic in vitro, haptenizes tissue proteins, and is immunogenic in rodents. The purpose of this pilot study was to determine whether SMX-NO, when administered to dogs, would lead to drug-tissue adducts, anti-drug antibodies, antioxidant depletion, or clinical evidence of drug hypersensitivity. Four dogs were randomized to one of four treatments: SMX-NO 1 mg/kg; SMX-NO 3 mg/kg; SMX-NO 10 mg/kg; or vehicle control. Dosing was by the intraperitoneal route, once daily for four consecutive days per week, for 2 weeks total, followed by a third week of observation. Following this, all dogs were challenged with trimethoprim-sulfamethoxazole, 25 mg/kg for 12 h for 2 weeks. No dog developed clinical or biochemical evidence of drug hypersensitivity. Plasma cysteine and leukocyte reduced glutathione were not depleted during dosing; however, ascorbate was significantly depleted by week 2 following SMX-NO at 10 mg/kg. Anti-SMX antibodies (IgG or IgM by ELISA) were not detected in any dogs at any time points. SMX-hemoglobin adducts were detected in the spleen in SMX-NO dosed dogs; however, these adducts were not accompanied by an immunologic or systemic response. The results of this pilot study indicate that SMX-NO dosing in dogs, using a dosing protocol shown to be immunogenic in other species, produces modest ascorbate depletion and hemoglobin adduct formation, but is insufficient to produce an immunologic response or a clinical syndrome of sulfonamide hypersensitivity in this susceptible species.
Collapse
Affiliation(s)
- Sidonie N Lavergne
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706-1102, USA
| | | | | | | | | |
Collapse
|