1
|
Frampton S, Smith R, Ferson L, Gibson J, Hollox EJ, Cragg MS, Strefford JC. Fc gamma receptors: Their evolution, genomic architecture, genetic variation, and impact on human disease. Immunol Rev 2024. [PMID: 39345014 DOI: 10.1111/imr.13401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Fc gamma receptors (FcγRs) are a family of receptors that bind IgG antibodies and interface at the junction of humoral and innate immunity. Precise regulation of receptor expression provides the necessary balance to achieve healthy immune homeostasis by establishing an appropriate immune threshold to limit autoimmunity but respond effectively to infection. The underlying genetics of the FCGR gene family are central to achieving this immune threshold by regulating affinity for IgG, signaling efficacy, and receptor expression. The FCGR gene locus was duplicated during evolution, retaining very high homology and resulting in a genomic region that is technically difficult to study. Here, we review the recent evolution of the gene family in mammals, its complexity and variation through copy number variation and single-nucleotide polymorphism, and impact of these on disease incidence, resolution, and therapeutic antibody efficacy. We also discuss the progress and limitations of current approaches to study the region and emphasize how new genomics technologies will likely resolve much of the current confusion in the field. This will lead to definitive conclusions on the impact of genetic variation within the FCGR gene locus on immune function and disease.
Collapse
Affiliation(s)
- Sarah Frampton
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Rosanna Smith
- Antibody and Vaccine Group, Faculty of Medicine, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Lili Ferson
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Jane Gibson
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Edward J Hollox
- Department of Genetics, Genomics and Cancer Sciences, College of Life Sciences, University of Leicester, Leicester, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Faculty of Medicine, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Jonathan C Strefford
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
2
|
Popa LG, Dumitras I, Giurcaneanu C, Berghi O, Radaschin DS, Vivisenco CI, Popescu MN, Beiu C. Mechanisms of Resistance to Rituximab Used for the Treatment of Autoimmune Blistering Diseases. Life (Basel) 2024; 14:1223. [PMID: 39459523 PMCID: PMC11508628 DOI: 10.3390/life14101223] [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: 08/05/2024] [Revised: 09/10/2024] [Accepted: 09/21/2024] [Indexed: 10/28/2024] Open
Abstract
Autoimmune blistering diseases represent a group of chronic severe, disabling, and potentially fatal disorders of the skin and/or mucous membranes, primarily mediated by pathogenic auto-antibodies. Despite their rarity, these diseases are associated with significant morbidity and mortality and profound negative impact on the patient's quality of life and impose a considerable economic burden. Rituximab, an anti-CD-20 monoclonal antibody, represents the first line of therapy for pemphigus, regardless of severity and a valuable off-label therapeutic alternative for subepidermal autoimmune blistering diseases as it ensures high rates of rapid, long-lasting complete remission. Nevertheless, disease recurrence is the rule, all patients requiring maintenance therapy with rituximab eventually. While innate resistance to rituximab in pemphigus patients is exceptional, acquired resistance is frequent and may develop even in patients with initial complete response to rituximab, representing a real challenge for physicians. We discuss the various resistance mechanisms and their complex interplay, as well as the numerous therapeutic alternatives that may be used to circumvent rituximab resistance. As no therapeutic measure is universally efficient, individualization of rituximab treatment regimen and tailored adjuvant therapies in refractory autoimmune blistering diseases are mandatory.
Collapse
Affiliation(s)
- Liliana Gabriela Popa
- Department of Dermatology, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, District 1, 020021 Bucharest, Romania
- Department of Dermatology, Elias Emergency University Hospital, 17 Marasti Bd., District 1, 011461 Bucharest, Romania
| | - Ioana Dumitras
- Department of Dermatology, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, District 1, 020021 Bucharest, Romania
| | - Calin Giurcaneanu
- Department of Dermatology, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, District 1, 020021 Bucharest, Romania
- Department of Dermatology, Elias Emergency University Hospital, 17 Marasti Bd., District 1, 011461 Bucharest, Romania
| | - Ovidiu Berghi
- Department of Allergy and Clinical Immunology, Colentina Clinical Hospital, 19-21 Stefan cel Mare Bd., District 2, 020125 Bucharest, Romania
| | - Diana Sabina Radaschin
- Department of Dermatology, Dunarea de Jos University of Medicine and Pharmacy, 25 Otelarilor Bd., 800008 Galati, Romania
| | - Cristina Iolanda Vivisenco
- Department of Paediatrics, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, District 1, 020021 Bucharest, Romania
- Department of Pediatrics, Grigore Alexandrescu Clinical Emergency Hospital for Children, 30-32 Iancu de Hunedoara Road, 011743 Bucharest, Romania
| | - Marius Nicolae Popescu
- Department of Physical and Rehabilitation Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, District 1, 020021 Bucharest, Romania
- Department of Physical and Rehabilitation Medicine, Dermatology Department, Elias Emergency University Hospital, 17 Marasti Bd., District 1, 011461 Bucharest, Romania
| | - Cristina Beiu
- Department of Dermatology, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, District 1, 020021 Bucharest, Romania
- Department of Dermatology, Elias Emergency University Hospital, 17 Marasti Bd., District 1, 011461 Bucharest, Romania
| |
Collapse
|
3
|
Al-Sofi RF, Bergmann MS, Nielsen CH, Andersen V, Skov L, Loft N. The Association between Genetics and Response to Treatment with Biologics in Patients with Psoriasis, Psoriatic Arthritis, Rheumatoid Arthritis, and Inflammatory Bowel Diseases: A Systematic Review and Meta-Analysis. Int J Mol Sci 2024; 25:5793. [PMID: 38891983 PMCID: PMC11171831 DOI: 10.3390/ijms25115793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Genetic biomarkers could potentially lower the risk of treatment failure in chronic inflammatory diseases (CID) like psoriasis, psoriatic arthritis (PsA), rheumatoid arthritis (RA), and inflammatory bowel disease (IBD). We performed a systematic review and meta-analysis assessing the association between single nucleotide polymorphisms (SNPs) and response to biologics. Odds ratio (OR) with 95% confidence interval (CI) meta-analyses were performed. In total, 185 studies examining 62,774 individuals were included. For the diseases combined, the minor allele of MYD88 (rs7744) was associated with good response to TNFi (OR: 1.24 [1.02-1.51], 6 studies, 3158 patients with psoriasis or RA) and the minor alleles of NLRP3 (rs4612666) (OR: 0.71 [0.58-0.87], 5 studies, 3819 patients with RA or IBD), TNF-308 (rs1800629) (OR: 0.71 [0.55-0.92], 25 studies, 4341 patients with psoriasis, RA, or IBD), FCGR3A (rs396991) (OR: 0.77 [0.65-0.93], 18 studies, 2562 patients with psoriasis, PsA, RA, or IBD), and TNF-238 (rs361525) (OR: 0.57 [0.34-0.96]), 7 studies, 818 patients with psoriasis, RA, or IBD) were associated with poor response to TNFi together or infliximab alone. Genetic variants in TNFα, NLRP3, MYD88, and FcRγ genes are associated with response to TNFi across several inflammatory diseases. Most other genetic variants associated with response were observed in a few studies, and further validation is needed.
Collapse
Affiliation(s)
- Rownaq Fares Al-Sofi
- Department of Dermatology and Allergy, Copenhagen University Hospital—Herlev and Gentofte, 1165 Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Mie Siewertsen Bergmann
- Department of Dermatology and Allergy, Copenhagen University Hospital—Herlev and Gentofte, 1165 Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Claus Henrik Nielsen
- Center for Rheumatology and Spine Diseases, Institute for Inflammation Research, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Vibeke Andersen
- Institute of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
- Molecular Diagnostics and Clinical Research Unit, Department of Internal Medicine, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Lone Skov
- Department of Dermatology and Allergy, Copenhagen University Hospital—Herlev and Gentofte, 1165 Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Nikolai Loft
- Department of Dermatology and Allergy, Copenhagen University Hospital—Herlev and Gentofte, 1165 Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| |
Collapse
|
4
|
Zhou Y, Guo R, Xia X, Jing S, Lu J, Ruan Z, Luo S, Huan X, Zhao C, Chang T, Xi J. A predictive nomogram for short-term outcomes of myasthenia gravis patients treated with low-dose rituximab. CNS Neurosci Ther 2024; 30:e14761. [PMID: 38739094 PMCID: PMC11090079 DOI: 10.1111/cns.14761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 04/08/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND This study aims to establish and validate a predictive nomogram for the short-term clinical outcomes of myasthenia gravis (MG) patients treated with low-dose rituximab. METHODS We retrospectively reviewed 108 patients who received rituximab of 600 mg every 6 months in Huashan Hospital and Tangdu Hospital. Of them, 76 patients from Huashan Hospital were included in the derivation cohort to develop the predictive nomogram, which was externally validated using 32 patients from Tangdu Hospital. The clinical response is defined as a ≥ 3 points decrease in QMG score within 6 months. Both clinical and genetic characteristics were included to screen predictors via multivariate logistic regression. Discrimination and calibration were measured by the area under the receiver operating characteristic curve (AUC-ROC) and Hosmer-Lemeshow test, respectively. RESULTS Disease duration (OR = 0.987, p = 0.032), positive anti-muscle-specific tyrosine kinase antibodies (OR = 19.8, p = 0.007), and genotypes in FCGR2A rs1801274 (AG: OR = 0.131, p = 0.024;GG:OR = 0.037, p = 0.010) were independently associated with clinical response of post-rituximab patients. The nomogram identified MG patients with clinical response with an AUC-ROC (95% CI) of 0.875 (0.798-0.952) in the derivation cohort and 0.741(0.501-0.982) in the validation cohort. Hosmer-Lemeshow test showed a good calibration (derivation: Chi-square = 3.181, p = 0.923; validation: Chi-square = 8.098, p = 0.424). CONCLUSIONS The nomogram achieved an optimal prediction of short-term outcomes in patients treated with low-dose rituximab.
Collapse
Affiliation(s)
- Yufan Zhou
- Department of Neurology, Huashan HospitalFudan UniversityShanghaiChina
- Huashan Rare Disease Center, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DiseasesShanghaiChina
| | - Rongjing Guo
- Department of Neurology, Tangdu HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Xingyu Xia
- Department of Neurology, Huashan HospitalFudan UniversityShanghaiChina
- Huashan Rare Disease Center, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DiseasesShanghaiChina
| | - Sisi Jing
- Department of Neurology, Banan HospitalChongqing Medical UniversityChongqingChina
| | - Jun Lu
- Department of Neurology, Huashan HospitalFudan UniversityShanghaiChina
- Huashan Rare Disease Center, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DiseasesShanghaiChina
| | - Zhe Ruan
- Department of Neurology, Tangdu HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Sushan Luo
- Department of Neurology, Huashan HospitalFudan UniversityShanghaiChina
- Huashan Rare Disease Center, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DiseasesShanghaiChina
| | - Xiao Huan
- Department of Neurology, Huashan HospitalFudan UniversityShanghaiChina
- Huashan Rare Disease Center, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DiseasesShanghaiChina
| | - Chongbo Zhao
- Department of Neurology, Huashan HospitalFudan UniversityShanghaiChina
- Huashan Rare Disease Center, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DiseasesShanghaiChina
| | - Ting Chang
- Department of Neurology, Tangdu HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Jianying Xi
- Department of Neurology, Huashan HospitalFudan UniversityShanghaiChina
- Huashan Rare Disease Center, Huashan HospitalFudan UniversityShanghaiChina
- National Center for Neurological DiseasesShanghaiChina
| |
Collapse
|
5
|
Lee YH, Song GG. Association between functional FCGR3A F158V and FCGR2A R131H polymorphisms and responsiveness to rituximab in patients with autoimmune diseases: a meta-analysis. THE PHARMACOGENOMICS JOURNAL 2023; 23:210-216. [PMID: 37149714 DOI: 10.1038/s41397-023-00308-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/29/2023] [Accepted: 04/17/2023] [Indexed: 05/08/2023]
Abstract
OBJECTIVES To investigate the association between the functional Fc gamma receptor 3 A (FCGR3A) V158F and FCGR2A R131H polymorphisms and rituximab therapy in patients with autoimmune diseases. METHODS We searched the Medline, Embase, and Cochrane databases for relevant articles. We conducted a meta-analysis of the association between FCGR3A V158F and FCGR2A R131H polymorphisms and responsiveness to rituximab in patients with autoimmune diseases. RESULTS Eleven studies, consisting of 661 responders and 267 non-responders for FCGR3A V158F polymorphism and 156 responders and 89 non-responders for FCGR2A R131H polymorphism, were included. The meta-analysis revealed a significant association between the FCGR3A V allele and responsiveness to rituximab (odds ratio [OR] = 1.600, 95% confidence interval [CI] = 1.268-2.018, P < 0.001). Furthermore, associations were found using the dominant and homozygous contrast models. Subgroup analysis showed an association between the FCGR3A V allele and responsiveness to rituximab in European, RA, ITP, small (<50) and large (≥50) groups, and short- (≤6 months) and long-term follow-up periods (≥6 months). These associations were also found in recessive, dominant or homozygous contrast models. Meta-analysis revealed no association between the FCGR2A R allele and responsiveness to rituximab (OR = 1.243, 95% CI = 0.825-1.873, P = 0.229). CONCLUSIONS We demonstrated that the FCGR3A F158V polymorphism is associated with better responsiveness to rituximab therapy in patients with autoimmune diseases, indicating that individuals carrying the FCGR3A V allele will likely respond better to rituximab. However, FCGR2A R131H polymorphism was not associated with better response to rituximab.
Collapse
Affiliation(s)
- Young Ho Lee
- Department of Rheumatology, Korea University Medicine, Seoul, Korea.
| | - Gwan Gyu Song
- Department of Rheumatology, Korea University Medicine, Seoul, Korea
| |
Collapse
|
6
|
Lee YH, Song GG. Association between the functional FCGR3A F158V and FCGR2A R131H polymorphisms and responsiveness to biologics in rheumatoid arthritis patients: A meta-analysis. Int J Rheum Dis 2023. [PMID: 37114884 DOI: 10.1111/1756-185x.14719] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 04/11/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVES This study aimed to investigate the association between functional Fc gamma receptor 3A (FCGR3A) V158F and FCGR2A R131H polymorphisms and biologic therapy in rheumatoid arthritis (RA) patients. METHODS We searched Medline, Embase, and Cochran databases for available articles. This study is a meta-analysis of the association between the FCGR3A V158F and FCGR2A R131H polymorphisms and their responsiveness to biologics in RA patients. RESULTS Seventeen studies involving RA patients with FCGR3A V158F (n = 1884) and FCGR2A R131H (n = 1118) polymorphisms were considered. This meta-analysis showed that the FCGR3A V allele was associated with responsiveness to rituximab (odds ratio [OR] = 1.431, 95% CI = 1.081-1.894, P = 0.012), but not with tumor necrosis factor (TNF) blockers, tocilizumab, or abatacept. A significant association was also found between the FCGR3A V158F polymorphism and responsiveness to biologics using the dominant-recessive model. Additionally, the FCGR3A V158F polymorphism was associated with responsiveness to TNF blockers in the homozygous contrast model. Meta-analysis revealed an association between the FCGR2A RR + RH genotype and responsiveness to biologics (OR = 1.385, 95% CI = 1.007-1.904, P = 0.045). CONCLUSIONS This meta-analysis demonstrates that FCGR3A V allele carriers show better responsiveness to rituximab, and FCGR2A R allele carriers may show a better response to biologics in RA treatment. Genotyping of these polymorphisms could be a useful tool to find associations with the responsiveness of personalized medicine to biologics.
Collapse
Affiliation(s)
- Young Ho Lee
- Department of Rheumatology, Korea University Medicine, Seoul, South Korea
| | - Gwan Gyu Song
- Department of Rheumatology, Korea University Medicine, Seoul, South Korea
| |
Collapse
|
7
|
Robinson JI, Md Yusof MY, Davies V, Wild D, Morgan M, Taylor JC, El-Sherbiny Y, Morris DL, Liu L, Rawstron AC, Buch MH, Plant D, Cordell HJ, Isaacs JD, Bruce IN, Emery P, Barton A, Vyse TJ, Barrett JH, Vital EM, Morgan AW. Comprehensive genetic and functional analyses of Fc gamma receptors influence on response to rituximab therapy for autoimmunity. EBioMedicine 2022; 86:104343. [PMID: 36371989 PMCID: PMC9663864 DOI: 10.1016/j.ebiom.2022.104343] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/07/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Rituximab is widely used to treat autoimmunity but clinical response varies. Efficacy is determined by the efficiency of B-cell depletion, which may depend on various Fc gamma receptor (FcγR)-dependent mechanisms. Study of FcγR is challenging due to the complexity of the FCGR genetic locus. We sought to assess the effect of FCGR variants on clinical response, B-cell depletion and NK-cell-mediated killing in rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). METHODS A longitudinal cohort study was conducted in 835 patients [RA = 573; SLE = 262]. Clinical outcome measures were two-component disease activity score in 28-joints (2C-DAS28CRP) for RA and British Isles Lupus Assessment Group (BILAG)-2004 major clinical response (MCR) for SLE at 6 months. B-cells were evaluated by highly-sensitive flow cytometry. Single nucleotide polymorphism and copy number variation for genes encoding five FcγRs were measured using multiplex ligation-dependent probe amplification. Ex vivo studies assessed NK-cell antibody-dependent cellular cytotoxicity (ADCC) and FcγR expression. FINDINGS In RA, carriage of FCGR3A-158V and increased FCGR3A-158V copies were associated with greater 2C-DAS28CRP response (adjusted for baseline 2C-DAS28CRP). In SLE, MCR was associated with increased FCGR3A-158V, OR 1.64 (95% CI 1.12-2.41) and FCGR2C-ORF OR 1.93 (95% CI 1.09-3.40) copies. 236/413 (57%) patients with B-cell data achieved complete depletion. Homozygosity for FCGR3A-158V and increased FCGR3A-158V copies were associated with complete depletion in combined analyses. FCGR3A genotype was associated with rituximab-induced ADCC, and increased NK-cell FcγRIIIa expression was associated with improved clinical response and depletion in vivo. Furthermore, disease status and concomitant therapies impacted both NK-cell FcγRIIIa expression and ADCC. INTERPRETATION FcγRIIIa is the major low affinity FcγR associated with rituximab response. Increased copies of the FCGR3A-158V allele (higher affinity for IgG1), influences clinical and biological responses to rituximab in autoimmunity. Enhancing FcγR-effector functions could improve the next generation of CD20-depleting therapies and genotyping may stratify patients for optimal treatment protocols. FUNDING Medical Research Council, National Institute for Health and Care Research, Versus Arthritis.
Collapse
Affiliation(s)
- James I Robinson
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK
| | - Md Yuzaiful Md Yusof
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK
| | - Vinny Davies
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK; School of Mathematics and Statistics, University of Glasgow, UK
| | - Dawn Wild
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK
| | - Michael Morgan
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK; Cancer Research UK Cambridge Institute, University of Cambridge, UK
| | - John C Taylor
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK
| | - Yasser El-Sherbiny
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, UK; Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - David L Morris
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, UK
| | - Lu Liu
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, UK
| | - Andy C Rawstron
- Haematological Malignancy Diagnostic Service, Leeds Teaching Hospitals NHS Trust, UK
| | - Maya H Buch
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK; Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and NIHR Manchester BRC, Manchester University NHS Foundation Trust, UK
| | - Darren Plant
- Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and NIHR Manchester BRC, Manchester University NHS Foundation Trust, UK
| | | | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University and Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, UK
| | - Ian N Bruce
- Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and NIHR Manchester BRC, Manchester University NHS Foundation Trust, UK
| | - Paul Emery
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK; NIHR Leeds Medtech and In vitro Diagnostics Co-operative, Leeds Teaching Hospitals NHS Trust, UK
| | - Anne Barton
- Versus Arthritis Centre for Genetics and Genomics, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and NIHR Manchester BRC, Manchester University NHS Foundation Trust, UK
| | - Timothy J Vyse
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, UK
| | - Jennifer H Barrett
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK
| | - Edward M Vital
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK
| | - Ann W Morgan
- School of Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, UK; NIHR Leeds Medtech and In vitro Diagnostics Co-operative, Leeds Teaching Hospitals NHS Trust, UK.
| |
Collapse
|
8
|
Genetic Variation among Pharmacogenes in the Sardinian Population. Int J Mol Sci 2022; 23:ijms231710058. [PMID: 36077453 PMCID: PMC9456055 DOI: 10.3390/ijms231710058] [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: 07/26/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
Pharmacogenetics (PGx) aims to identify the genetic factors that determine inter-individual differences in response to drug treatment maximizing efficacy while decreasing the risk of adverse events. Estimating the prevalence of PGx variants involved in drug response, is a critical preparatory step for large-scale implementation of a personalized medicine program in a target population. Here, we profiled pharmacogenetic variation in fourteen clinically relevant genes in a representative sample set of 1577 unrelated sequenced Sardinians, an ancient island population that accounts for genetic variation in Europe as a whole, and, at the same time is enriched in genetic variants that are very rare elsewhere. To this end, we used PGxPOP, a PGx allele caller based on the guidelines created by the Clinical Pharmacogenetics Implementation Consortium (CPIC), to identify the main phenotypes associated with the PGx alleles most represented in Sardinians. We estimated that 99.43% of Sardinian individuals might potentially respond atypically to at least one drug, that on average each individual is expected to have an abnormal response to about 17 drugs, and that for 27 drugs the fraction of the population at risk of atypical responses to therapy is more than 40%. Finally, we identified 174 pharmacogenetic variants for which the minor allele frequency was at least 10% higher among Sardinians as compared to other European populations, a fact that may contribute to substantial interpopulation variability in drug response phenotypes. This study provides baseline information for further large-scale pharmacogenomic investigations in the Sardinian population and underlines the importance of PGx characterization of diverse European populations, such as Sardinians.
Collapse
|
9
|
Gonzalez JC, Chakraborty S, Thulin NK, Wang TT. Heterogeneity in IgG-CD16 signaling in infectious disease outcomes. Immunol Rev 2022; 309:64-74. [PMID: 35781671 PMCID: PMC9539944 DOI: 10.1111/imr.13109] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In this review, we discuss how IgG antibodies can modulate inflammatory signaling during viral infections with a focus on CD16a-mediated functions. We describe the structural heterogeneity of IgG antibody ligands, including subclass and glycosylation that impact binding by and downstream activity of CD16a, as well as the heterogeneity of CD16a itself, including allele and expression density. While inflammation is a mechanism required for immune homeostasis and resolution of acute infections, we focus here on two infectious diseases that are driven by pathogenic inflammatory responses during infection. Specifically, we review and discuss the evolving body of literature showing that afucosylated IgG immune complex signaling through CD16a contributes to the overwhelming inflammatory response that is central to the pathogenesis of severe forms of dengue disease and coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Joseph C. Gonzalez
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA,Program in ImmunologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Saborni Chakraborty
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA
| | - Natalie K. Thulin
- Department of ImmunologyUniversity of WashingtonSeattleWashingtonUSA
| | - Taia T. Wang
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA,Program in ImmunologyStanford University School of MedicineStanfordCaliforniaUSA,Department of Microbiology and ImmunologyStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
10
|
Lim SH, Kim K, Choi CI. Pharmacogenomics of Monoclonal Antibodies for the Treatment of Rheumatoid Arthritis. J Pers Med 2022; 12:jpm12081265. [PMID: 36013214 PMCID: PMC9410311 DOI: 10.3390/jpm12081265] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Precision medicine refers to a highly individualized and personalized approach to patient care. Pharmacogenomics is the study of how an individual’s genomic profile affects their drug response, enabling stable and effective drug selection, minimizing side effects, and maximizing therapeutic efficacy. Rheumatoid arthritis (RA) is an autoimmune disease that causes chronic inflammation in the joints. It mainly starts in peripheral joints, such as the hands and feet, and progresses to large joints, which causes joint deformation and bone damage due to inflammation of the synovial membrane. Here, we review various pharmacogenetic studies investigating the association between clinical response to monoclonal antibody therapy and their target genetic polymorphisms. Numerous papers have reported that some single nucleotide polymorphisms (SNPs) are related to the therapeutic response of several monoclonal antibody drugs including adalimumab, infliximab, rituximab, and tocilizumab, which target tumor necrosis factor (TNF), CD20 of B-cells, and interleukin (IL)-6. Additionally, there are some pharmacogenomic studies reporting on the association between the clinical response of monoclonal antibodies having various mechanisms, such as IL-1, IL-17, IL-23, granulocyte-macrophage colony-stimulating factor (GM-CSF) and the receptor activator of nuclear factor-kappa B (RANK) inhibition. Biological therapies are currently prescribed on a “trial and error” basis for RA patients. If appropriate drug treatment is not started early, joints may deform, and long-term treatment outcomes may worsen. Pharmacogenomic approaches that predict therapeutic responses for RA patients have the potential to significantly improve patient quality of life and reduce treatment costs.
Collapse
Affiliation(s)
- Sung Ho Lim
- Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea;
| | - Khangyoo Kim
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea;
| | - Chang-Ik Choi
- Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea;
- Correspondence: ; Tel.: +82-31-961-5230
| |
Collapse
|
11
|
Aluko A, Ranganathan P. Pharmacogenetics of Drug Therapies in Rheumatoid Arthritis. Methods Mol Biol 2022; 2547:527-567. [PMID: 36068476 DOI: 10.1007/978-1-0716-2573-6_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic inflammatory disorder that can lead to severe joint damage and is often associated with a high morbidity and disability. Disease-modifying anti-rheumatic drugs (DMARDs) are the mainstay of treatment in RA. DMARDs not only relieve the clinical signs and symptoms of RA but also inhibit the radiographic progression of disease and reduce the effects of chronic systemic inflammation. Since the introduction of biologic DMARDs in the late 1990s, the therapeutic range of options for the management of RA has significantly expanded. However, patients' response to these agents is not uniform with considerable variability in both efficacy and toxicity. There are no reliable means of predicting an individual patient's response to a given DMARD prior to initiation of therapy. In this chapter, the current published literature on the pharmacogenetics of traditional DMARDS and the newer biologic DMARDs in RA is highlighted. Pharmacogenetics may help individualize drug therapy in patients with RA by providing reliable biomarkers to predict medication toxicity and efficacy.
Collapse
Affiliation(s)
- Atinuke Aluko
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Prabha Ranganathan
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
12
|
Wang Z, Huang J, Xie D, He D, Lu A, Liang C. Toward Overcoming Treatment Failure in Rheumatoid Arthritis. Front Immunol 2021; 12:755844. [PMID: 35003068 PMCID: PMC8732378 DOI: 10.3389/fimmu.2021.755844] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder characterized by inflammation and bone erosion. The exact mechanism of RA is still unknown, but various immune cytokines, signaling pathways and effector cells are involved. Disease-modifying antirheumatic drugs (DMARDs) are commonly used in RA treatment and classified into different categories. Nevertheless, RA treatment is based on a "trial-and-error" approach, and a substantial proportion of patients show failed therapy for each DMARD. Over the past decades, great efforts have been made to overcome treatment failure, including identification of biomarkers, exploration of the reasons for loss of efficacy, development of sequential or combinational DMARDs strategies and approval of new DMARDs. Here, we summarize these efforts, which would provide valuable insights for accurate RA clinical medication. While gratifying, researchers realize that these efforts are still far from enough to recommend specific DMARDs for individual patients. Precision medicine is an emerging medical model that proposes a highly individualized and tailored approach for disease management. In this review, we also discuss the potential of precision medicine for overcoming RA treatment failure, with the introduction of various cutting-edge technologies and big data.
Collapse
Affiliation(s)
- Zhuqian Wang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Jie Huang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Duoli Xie
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Dongyi He
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Chao Liang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
13
|
Hashimoto T, Yoshida K, Hashiramoto A, Matsui K. Cell-Free DNA in Rheumatoid Arthritis. Int J Mol Sci 2021; 22:8941. [PMID: 34445645 PMCID: PMC8396202 DOI: 10.3390/ijms22168941] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023] Open
Abstract
Endogenous DNA derived from the nuclei or mitochondria is released into the bloodstream following cell damage or death. Extracellular DNA, called cell-free DNA (cfDNA), is associated with various pathological conditions. Recently, multiple aspects of cfDNA have been assessed, including cfDNA levels, integrity, methylation, and mutations. Rheumatoid arthritis (RA) is the most common form of autoimmune arthritis, and treatment of RA has highly varied outcomes. cfDNA in patients with RA is elevated in peripheral blood and synovial fluid and is associated with disease activity. Profiling of cfDNA in patients with RA may then be utilized in various aspects of clinical practice, such as the prediction of prognosis and treatment responses; monitoring disease state; and as a diagnostic marker. In this review, we discuss cfDNA in patients with RA, particularly the sources of cfDNA and the correlation of cfDNA with RA pathogenesis. We also highlight the potential of analyzing cfDNA profiles to guide individualized treatment approaches for RA.
Collapse
Affiliation(s)
- Teppei Hashimoto
- Division of Diabetes, Endocrinology and Clinical Immunology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya 6638501, Japan;
| | - Kohsuke Yoshida
- Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe 6540142, Japan; (K.Y.); (A.H.)
| | - Akira Hashiramoto
- Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe 6540142, Japan; (K.Y.); (A.H.)
| | - Kiyoshi Matsui
- Division of Diabetes, Endocrinology and Clinical Immunology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya 6638501, Japan;
| |
Collapse
|
14
|
Márquez Pete N, Maldonado Montoro MDM, Pérez Ramírez C, Martínez Martínez F, Martínez de la Plata JE, Daddaoua A, Jiménez Morales A. Influence of the FCGR2A rs1801274 and FCGR3A rs396991 Polymorphisms on Response to Abatacept in Patients with Rheumatoid Arthritis. J Pers Med 2021; 11:jpm11060573. [PMID: 34207385 PMCID: PMC8233911 DOI: 10.3390/jpm11060573] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 12/13/2022] Open
Abstract
Abatacept (ABA) is an immunosuppressant indicated for treatment of rheumatoid arthritis (RA). Effectiveness might be influenced by clinical RA variants and single-nucleotide polymorphisms (SNPs) in genes encoding protein FCGR2A (His131Arg) and FCGR3A (Phe158Val) involved in pharmacokinetics of ABA. An observational cohort study was conducted in 120 RA Caucasian patients treated with ABA for 6 and 12 months. Patients with the FCGR2A rs1801274-AA genotype (FCGR2A-p.131His) showed a better EULAR response (OR = 2.43; 95% CI = 1.01-5.92) at 12 months and low disease activity (LDA) at 6 months (OR = 3.16; 95% CI = 1.19-8.66) and 12 months (OR = 6.62; 95% CI = 1.25-46.89) of treatment with ABA. A tendency was observed towards an association between the FCGR3A rs396991-A allele (FCGR3A-p.158Phe) and better therapeutic response to ABA after 12 months of treatment (p = 0.078). Moreover, we found a significant association between the low-affinity FCGR2A/FCGR3A haplotypes variable and LDA after 12 months of ABA treatment (OR = 1.59; 95% CI = 1.01-2.58). The clinical variables associated with better response to ABA were lower age at starting ABA (OR = 1.06; 95% CI = 1.02-1.11) and greater duration of ABA treatment (OR = 1.02; 95% CI = 1.01-1.04), lower duration of previous biological therapies (OR = 0.99; 95% CI = 0.98-0.99), non-administration of concomitant disease-modifying antirheumatic drugs (DMARDs) (OR = 24.53; 95% CI = 3.46-523.80), non-use of concomitant glucocorticoids (OR = 0.12; 95% CI = 0.02-0.47), monotherapy (OR = 19.22; 95% CI = 2.05-343.00), lower initial patient's visual analogue scale (PVAS) value (OR = 0.95; 95% CI = 0.92-0.97), and lower baseline ESR (OR = 0.92; 95% CI = 0.87-0.97). This study showed that high-affinity FCGR2A-p.131His variant, low-affinity FCGR3A-p.158Phe variant, and combined use of FCGR2A/FCGR3A genetic variations could affect ABA effectiveness. Further studies will be required to confirm these results.
Collapse
Affiliation(s)
- Noelia Márquez Pete
- Pharmacy Service, Pharmacogenetics Unit, University Hospital Virgen de las Nieves, 18014 Granada, Spain; (N.M.P.); (A.J.M.)
| | | | - Cristina Pérez Ramírez
- Pharmacy Service, Pharmacogenetics Unit, University Hospital Virgen de las Nieves, 18014 Granada, Spain; (N.M.P.); (A.J.M.)
- Center of Biomedical Research, Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Avda. del Conocimiento s/n., 18016 Granada, Spain
- Correspondence:
| | - Fernando Martínez Martínez
- Department of Pharmacy and Pharmaceutical Technology, Social and Legal Assistance Pharmacy Section, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain;
| | | | - Abdelali Daddaoua
- Department of Biochemistry, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain;
| | - Alberto Jiménez Morales
- Pharmacy Service, Pharmacogenetics Unit, University Hospital Virgen de las Nieves, 18014 Granada, Spain; (N.M.P.); (A.J.M.)
| |
Collapse
|
15
|
Nagafuchi H, Goto Y, Suzuki S, Sakurai K, Imamura M, Suzuki T, Yamasaki Y, Shibata T, Kawahata K. Rheumatoid arthritis relapse in patients with other iatrogenic immunodeficiency-associated lymphoproliferative disorders and its treatment. Mod Rheumatol 2021; 31:1087-1093. [PMID: 33491519 DOI: 10.1080/14397595.2021.1879367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) in patients undergoing immunosuppressive therapy (IS) is sometimes involved with other iatrogenic immunodeficiency-associated lymphoproliferative disorders (LPD). We aimed to clarify the effects of LPD treatment on RA and the current status of RA treatment options after LPD onset and subsequent IS withdrawal. METHODS We retrospectively analyzed data of patients who had RA with LPD and examined the relationship between LPD course and RA treatment as well as that between RA relapse and LPD treatment. RESULTS LPD patients were categorized into two groups: patients who regressed spontaneously (n = 19) and those who needed chemotherapy (n = 12). The chemotherapy group had significantly less RA relapse than the spontaneous regression group (p = .041). RA almost relapsed early in the spontaneous regression group and needed treatment for RA. Chemotherapy with rituximab prevented long-term RA relapse, and RA did not relapse for long even after rituximab monotherapy. The total dose of rituximab in monotherapy correlated with the time to RA relapse. Six patients with RA relapse received biologics and had no LPD relapse for more than 1 year. CONCLUSIONS Rituximab in chemotherapy for LPD may help prevent RA relapse with LPD. Large-scale studies are required in the future for verification.
Collapse
Affiliation(s)
- Hiroko Nagafuchi
- Division of Rheumatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yutaka Goto
- Division of Rheumatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Shotaro Suzuki
- Division of Rheumatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Keiichi Sakurai
- Division of Rheumatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Mitsuru Imamura
- Division of Rheumatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takeshi Suzuki
- Division of Rheumatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yoshioki Yamasaki
- Division of Rheumatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Tomohiko Shibata
- Division of Rheumatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kimito Kawahata
- Division of Rheumatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
16
|
Dedmon LE. The genetics of rheumatoid arthritis. Rheumatology (Oxford) 2021; 59:2661-2670. [PMID: 32638005 DOI: 10.1093/rheumatology/keaa232] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/03/2020] [Indexed: 12/14/2022] Open
Abstract
RA is a chronic systemic inflammatory disease that primarily affects the small joints of the hands and feet, and results in a mean reduction in life expectancy of 3-10 years. RA is a multigene disorder with a substantial genetic component and a heritability estimate of 60%. Large-scale Genome-Wide Association Studies (GWAS) and meta-analyses have revealed common disease-associated variants in the population that may contribute cumulatively to RA pathogenesis. This review identifies the most significant genetic variants associated with RA susceptibility to date, with particular focus on the contribution of the HLA class II genes across different ethnic groups. Also discussed are the potential applications of pharmacogenomics to RA management by identifying polymorphisms associated with variation in treatment response or toxicity. The use of genetic variants to guide treatment strategy has the potential to not only reduce National Health Service costs, but also drastically improve patient experience and quality of life.
Collapse
|
17
|
The growing role of precision medicine for the treatment of autoimmune diseases; results of a systematic review of literature and Experts' Consensus. Autoimmun Rev 2020; 20:102738. [PMID: 33326854 DOI: 10.1016/j.autrev.2020.102738] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Autoimmune diseases (AIDs) share similar serological, clinical, and radiological findings, but, behind these common features, there are different pathogenic mechanisms, immune cells dysfunctions, and targeted organs. In this context, multiple lines of evidence suggest the application of precision medicine principles to AIDs to reduce the treatment failure. Precision medicine refers to the tailoring of therapeutic strategies to the individual characteristics of each patient, thus it could be a new approach for management of AIDS which considers individual variability in genes, environmental exposure, and lifestyle. Precision medicine would also assist physicians in choosing the right treatment, the best timing of administration, consequently trying to maximize drug efficacy, and, possibly, reducing adverse events. In this work, the growing body of evidence is summarized regarding the predictive factors for drug response in patients with AIDs, applying the precision medicine principles to provide high-quality evidence for therapeutic opportunities in improving the management of these patients.
Collapse
|
18
|
Zhong M, van der Walt A, Campagna MP, Stankovich J, Butzkueven H, Jokubaitis V. The Pharmacogenetics of Rituximab: Potential Implications for Anti-CD20 Therapies in Multiple Sclerosis. Neurotherapeutics 2020; 17:1768-1784. [PMID: 33058021 PMCID: PMC7851267 DOI: 10.1007/s13311-020-00950-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
There are a broad range of disease-modifying therapies (DMTs) available in relapsing-remitting multiple sclerosis (RRMS), but limited biomarkers exist to personalise DMT choice. All DMTs, including monoclonal antibodies such as rituximab and ocrelizumab, are effective in preventing relapses and preserving neurological function in MS. However, each agent harbours its own risk of therapeutic failure or adverse events. Pharmacogenetics, the study of the effects of genetic variation on therapeutic response or adverse events, could improve the precision of DMT selection. Pharmacogenetic studies of rituximab in MS patients are lacking, but pharmacogenetic markers in other rituximab-treated autoimmune conditions have been identified. This review will outline the wider implications of pharmacogenetics and the mechanisms of anti-CD20 agents in MS. We explore the non-MS rituximab literature to characterise pharmacogenetic variants that could be of prognostic relevance in those receiving rituximab, ocrelizumab or other monoclonal antibodies for MS.
Collapse
Affiliation(s)
- Michael Zhong
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia.
- Department of Neurology, Alfred Health, Level 6, Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia.
| | - Anneke van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
- Department of Neurology, Alfred Health, Level 6, Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia
| | - Maria Pia Campagna
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Jim Stankovich
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
- Department of Neurology, Alfred Health, Level 6, Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
- Department of Neurology, Alfred Health, Level 6, Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia
| |
Collapse
|
19
|
Pallio G, Mannino F, Irrera N, Eid AH, Squadrito F, Bitto A. Polymorphisms Involved in Response to Biological Agents Used in Rheumatoid Arthritis. Biomolecules 2020; 10:biom10091203. [PMID: 32825059 PMCID: PMC7565539 DOI: 10.3390/biom10091203] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/11/2020] [Accepted: 08/15/2020] [Indexed: 12/14/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic disease that leads to joint destruction. During the last decade, the therapy of RA has been principally based on biological drugs. Although the efficacy of biological therapy has been established, patients demonstrated a high heterogeneity in clinical response to treatment. Several genetic polymorphisms play a part in the different response to biological drugs. This review summarizes the pharmacogenetics of biological agents approved for clinical RA treatment. We reviewed PubMed papers published over the past 20 years (2000-2020), inserting as the search term "rheumatoid arthritis and polymorphisms". Despite some studies showing important correlations between genetic polymorphisms and response to biological therapy in RA patients, most of these findings are still lacking and inconsistent. The personalized treatment according to a pharmacogenetics approach is promising but the available pharmacogenetics data on biological treatment in RA are not adequate and reliable to recommend pharmacogenetic tests before starting biological therapy in RA patients.
Collapse
Affiliation(s)
- Giovanni Pallio
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (G.P.); (F.M.); (N.I.); (F.S.)
| | - Federica Mannino
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (G.P.); (F.M.); (N.I.); (F.S.)
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (G.P.); (F.M.); (N.I.); (F.S.)
| | - Ali H. Eid
- Department of Pharmacology and Toxicology, American University of Beirut, Bliss Street, Riad El Solh, 1107-2020 Beirut, Lebanon;
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (G.P.); (F.M.); (N.I.); (F.S.)
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (G.P.); (F.M.); (N.I.); (F.S.)
- Correspondence: ; Tel.: +39-090-2213086; Fax: +39-090-2213300
| |
Collapse
|
20
|
Kunchok A, Malpas C, Nytrova P, Havrdova EK, Alroughani R, Terzi M, Yamout B, Hor JY, Karabudak R, Boz C, Ozakbas S, Olascoaga J, Simo M, Granella F, Patti F, McCombe P, Csepany T, Singhal B, Bergamaschi R, Fragoso Y, Al-Harbi T, Turkoglu R, Lechner-Scott J, Laureys G, Oreja-Guevara C, Pucci E, Sola P, Ferraro D, Altintas A, Soysal A, Vucic S, Grand'Maison F, Izquierdo G, Eichau S, Lugaresi A, Onofrj M, Trojano M, Marriott M, Butzkueven H, Kister I, Kalincik T. Clinical and therapeutic predictors of disease outcomes in AQP4-IgG+ neuromyelitis optica spectrum disorder. Mult Scler Relat Disord 2020; 38:101868. [DOI: 10.1016/j.msard.2019.101868] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/22/2019] [Accepted: 11/23/2019] [Indexed: 10/25/2022]
|
21
|
Machaj F, Rosik J, Szostak B, Pawlik A. The evolution in our understanding of the genetics of rheumatoid arthritis and the impact on novel drug discovery. Expert Opin Drug Discov 2019; 15:85-99. [PMID: 31661990 DOI: 10.1080/17460441.2020.1682992] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Rheumatoid arthritis (RA) is an autoimmune disease that is characterized by chronic inflammation of the joints and affects 1% of the population. Polymorphisms of genes that encode proteins that primarily participate in inflammation may influence RA occurrence or become useful biomarkers for certain types of anti-rheumatic treatment.Areas covered: The authors summarize the recent progress in our understanding of the genetics of RA. In the last few years, multiple variants of genes that are associated with RA risk have been identified. The development of new technologies and the detection of new potential therapeutic targets that contribute to novel drug discovery are also described.Expert opinion: There is still the need to search for new genes which may be a potential target for RA therapy. The challenge is to develop appropriate strategies for achieving insight into the molecular pathways involved in RA pathogenesis. Understanding the genetics, immunogenetics, epigenetics and immunology of RA could help to identify new targets for RA therapy. The development of new technologies has enabled the detection of a number of new genes, particularly genes associated with proinflammatory cytokines and chemokines, B- and T-cell activation pathways, signal transducers and transcriptional activators, which might be potential therapeutic targets in RA.
Collapse
Affiliation(s)
- Filip Machaj
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Jakub Rosik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
22
|
Tavakolpour S, Alesaeidi S, Darvishi M, GhasemiAdl M, Darabi-Monadi S, Akhlaghdoust M, Elikaei Behjati S, Jafarieh A. A comprehensive review of rituximab therapy in rheumatoid arthritis patients. Clin Rheumatol 2019; 38:2977-2994. [PMID: 31367943 DOI: 10.1007/s10067-019-04699-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022]
Abstract
Rituximab (RTX) is an approved treatment for rheumatoid arthritis (RA) patients that do not respond adequately to disease-modifying antirheumatic drugs. However, different new concerns, such as efficacy, optimum dose, safety issues, prediction of response to RTX, and pregnancy outcomes have attracted a lot of attention. The PubMed database was systematically reviewed for the last published articles, new findings, and controversial issues regarding RTX therapy in RA using "Rheumatoid arthritis" AND "rituximab" keywords, last updated on June 18, 2019. From 1812 initial recorders, 162 studies met the criteria. Regarding the optimum dose, low-dose RTX therapy (2 × 500 mg) seems as effective as standard dose (2 × 1000 mg), safer, and more cost-effective. The most common reported safety challenges included de novo infections, false negative serologic tests of viral infections, reactivation of chronic infections, interfering with vaccination outcome, and development of de novo psoriasis. Other less reported side effects are infusion reactions, nervous system disorders, and gastrointestinal disorders. Lower exposure to other biologics, presence of some serological markers (e.g., anti-RF, anti-CCP, IL-33, ESR), specific variations in FCGR3A, FCGR2A, TGFβ1, IL6, IRF5, BAFF genes, and also EBV-positivity could be used to predict response to RTX. Although there is no evidence of the teratogenic effect of RTX, it is recommended that women do not expose themselves to RTX at least 6 months before the conception. Only a reversible reduction of B cell-count in the offspring may be the pregnancy-related outcome. Although RTX is an effective therapeutic option for RA, more studies on optimum doses, prevention of RTX-related side effects, prediction of RTX response, and safety during the pregnancy are required.
Collapse
Affiliation(s)
- Soheil Tavakolpour
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Rheumatology and Internal Medicine, Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Samira Alesaeidi
- Rheumatology and Internal Medicine, Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Darvishi
- Infectious Diseases and Tropical Medicine Research Center (IDTMRC), department of aerospace and subaquatic medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mojtaba GhasemiAdl
- Rheumatology and Internal Medicine, Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Meisam Akhlaghdoust
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Arash Jafarieh
- Amir'Alam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Swart M, Stansberry WM, Pratt VM, Medeiros EB, Kiel PJ, Shen F, Schneider BP, Skaar TC. Analytical Validation of Variants to Aid in Genotype-Guided Therapy for Oncology. J Mol Diagn 2019; 21:491-502. [PMID: 30794985 DOI: 10.1016/j.jmoldx.2019.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/30/2018] [Accepted: 01/29/2019] [Indexed: 10/27/2022] Open
Abstract
The Clinical Laboratory Improvement Amendments of 1988 require that pharmacogenetic genotyping methods need to be established according to technical standards and laboratory practice guidelines before testing can be offered to patients. Testing methods for variants in ABCB1, CBR3, COMT, CYP3A7, C8ORF34, FCGR2A, FCGR3A, HAS3, NT5C2, NUDT15, SBF2, SEMA3C, SLC16A5, SLC28A3, SOD2, TLR4, and TPMT were validated in a Clinical Laboratory Improvement Amendments-accredited laboratory. Because no known reference materials were available, existing DNA samples were used for the analytical validation studies. Pharmacogenetic testing methods developed here were shown to be accurate and 100% analytically sensitive and specific. Other Clinical Laboratory Improvement Amendments-accredited laboratories interested in offering pharmacogenetic testing for these genetic variants, related to genotype-guided therapy for oncology, could use these publicly available samples as reference materials when developing and validating new genetic tests or refining current assays.
Collapse
Affiliation(s)
- Marelize Swart
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Wesley M Stansberry
- Department of Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Victoria M Pratt
- Department of Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana.
| | - Elizabeth B Medeiros
- Department of Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Patrick J Kiel
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Fei Shen
- Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Bryan P Schneider
- Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Todd C Skaar
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
24
|
Jiménez Morales A, Maldonado-Montoro M, Martínez de la Plata JE, Pérez Ramírez C, Daddaoua A, Alarcón Payer C, Expósito Ruiz M, García Collado C. FCGR2A/FCGR3A Gene Polymorphisms and Clinical Variables as Predictors of Response to Tocilizumab and Rituximab in Patients With Rheumatoid Arthritis. J Clin Pharmacol 2018; 59:517-531. [PMID: 30457672 DOI: 10.1002/jcph.1341] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/25/2018] [Indexed: 12/18/2022]
Abstract
We evaluated the influence of clinical, biochemical, and genetic factors on response in 142 patients diagnosed with rheumatoid arthritis, of whom 87 patients were treated with tocilizumab (61.26%) and 55 patients were treated with rituximab (38.7%;) according to the variables European League Against Rheumatism (EULAR) response, remission, low disease activity, and improvement in Disease Activity Score, 28 joints (DAS28) at 6, 12, and 18 months. A retrospective prospective cohort study was conducted. Patients carrying the FCGR3A rs396991-TT genotype treated with tocilizumab showed higher EULAR response (OR, 5.075; 95%CI, 1.20-21.33; P = .027) at 12 months, those who were naive for biological disease-modifying antirheumatic drugs (bDMARDs) at the beginning of treatment showed satisfactory EULAR response, higher remission, and greater improvement in DAS28 at 6 months. Younger age at start of tocilizumab treatment was associated with satisfactory EULAR response at 18 months and greater remission at 6 and 18 months. Subcutaneous tocilizumab administration was associated with higher remission at 6 months and improved low disease activity rate at 12 months. In patients treated with rituximab, carriers of the FCGR2A rs1801274-TT genotype had higher EULAR response at 6 months (OR, 4.861; 95%CI, 1.11-21.12; P = .035), 12 months (OR, 4.667; p = 0.066, 95%CI, 0.90-24.12; P = .066), and 18 months (OR, 2.487; 95%CI, 0.35-17.31; P = .357), higher remission (OR: 10.625; p = 0.044, CI95% : 1.07, 105.47) at 6 months, and greater improvement in DAS28 at 12 months (B = 0.782; 95%CI, -0.15 to 1.71; P = .098) and 18 months (B = 1.414; 95%CI, 0.19-2.63; P = .025). The FCGR3A rs396991-G allele was associated with improved low disease activity rate (OR, 4.904; 95%CI, 0.84-28.48; P = .077) and greater improvement in DAS28 (B = -1.083; 95%CI, -1.98 to -0.18; P = .021) at 18 months. Patients with a lower number of previous biological therapies had higher remission at 12 months. We suggest that the FCGR3A rs396991-TT genotype, higher baseline value of DAS28, subcutaneous tocilizumab administration, younger age at the beginning of treatment, and being bDMARD naive are associated with better response to tocilizumab. In patients treated with rituximab, we found better response in those patients with the FCGR2A rs1801274-TT genotype, the FCGR3A rs396991-G allele, and lower number of previous biological therapies.
Collapse
Affiliation(s)
- Alberto Jiménez Morales
- Pharmacy Service, UGC Provincial de Farmacia de Granada, University Hospital Virgen de las Nieves, Granada, Spain
| | - Mar Maldonado-Montoro
- Pharmacy Service, UGC Provincial de Farmacia de Granada, University Hospital Virgen de las Nieves, Granada, Spain
| | | | - Cristina Pérez Ramírez
- Pharmacy Service, UGC Provincial de Farmacia de Granada, University Hospital Virgen de las Nieves, Granada, Spain.,Department of Biochemistry, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Abdelali Daddaoua
- Department of Biochemistry, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Carolina Alarcón Payer
- Pharmacy Service, UGC Provincial de Farmacia de Granada, University Hospital Virgen de las Nieves, Granada, Spain
| | - Manuela Expósito Ruiz
- Fundación Pública Andaluza para la Investigación Biosanitaria de Andalucía Oriental "Alejandro Otero" (FIBAO), University Hospital Virgen de las Nieves, Granada, Spain
| | - Carlos García Collado
- Pharmacy Service, UGC Provincial de Farmacia de Granada, University Hospital Virgen de las Nieves, Granada, Spain
| |
Collapse
|
25
|
van Vollenhoven RF. Genotypes, phenotypes and treatment with immunomodulators in the rheumatic diseases. J Intern Med 2018; 284:228-239. [PMID: 29908080 DOI: 10.1111/joim.12800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The autoimmune rheumatological diseases rheumatoid arthritis (RA), spondyloarthritis (SpA) and systemic lupus erythematosus (SLE) are treated with conventional immunosuppressive agents and with modern biological immunomodulators. The latter group of medications have brought about a major change in our ability to control RA and SpA, with more modest results for SLE. The biologicals are very specific in their mechanisms of action, targeting one specific cytokine or one particular cellular marker. Because of this, their efficacy can readily be linked to a single immunomodulatory mechanism. This observation has fuelled hopes that the efficacy of these agents can be predicted at the individual level based on the patient's genetic predisposition, immunological profile or disease phenotype. Whilst the biologic therapies have improved the prospects for patients with these diseases very significantly, the hope that they could be targeted to the patient in an individualized manner has not completely born fruit. In this review, I will argue that we are witnessing important progress in this field, and that justified hope exists for true advances in precision medicine in the autoimmune diseases in the coming years.
Collapse
Affiliation(s)
- R F van Vollenhoven
- The Amsterdam Rheumatology and Immunology Center ARC, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Trend S, Jones AP, Cha L, Byrne SN, Geldenhuys S, Fabis-Pedrini MJ, Carroll WM, Cole JM, Booth DR, Lucas RM, Kermode AG, French MA, Hart PH. Higher Serum Immunoglobulin G3 Levels May Predict the Development of Multiple Sclerosis in Individuals With Clinically Isolated Syndrome. Front Immunol 2018; 9:1590. [PMID: 30057580 PMCID: PMC6053531 DOI: 10.3389/fimmu.2018.01590] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/27/2018] [Indexed: 11/13/2022] Open
Abstract
Clinically isolated syndrome (CIS) is a first episode of neurological symptoms that may precede a diagnosis of multiple sclerosis (MS). Therefore, studying individuals with CIS may lead to breakthroughs in understanding the development and pathogenesis of MS. In this study, serum levels of immunoglobulin (Ig)G, IgA, IgM, and IgG1–4 were measured in 20 people with CIS and compared with those in 10 healthy controls (HC) and 8 people with MS. Serum Ig levels in individuals with CIS were compared with (a) the time to their conversion from CIS to MS, (b) serum levels of antibodies to Epstein–Barr virus, (c) frequencies of T regulatory (Treg), T follicular regulatory (Tfr), and B cell subsets, and (d) Treg/Tfr expression of Helios. Serum IgG, IgM, and IgG2 levels were significantly lower in people with CIS than HC, and IgG, IgM, and IgG1 levels were significantly lower in people with CIS than MS. After adjusting for age, sex, and serum 25(OH) vitamin D3 [25(OH)D] levels, CIS was associated with lower serum levels of IgG and IgG2 compared with HC (p = 0.001 and p < 0.001, respectively). People with MS had lower IgG2 levels (p < 0.001) and IgG2 proportions (%IgG; p = 0.007) compared with HC. After adjusting for age, sex, and 25(OH)D, these outcomes remained, in addition to lower serum IgA levels (p = 0.01) and increased IgG3 levels (p = 0.053) in people with MS compared with HC. Furthermore, serum from people with MS had increased proportions of IgG1 and IgG3 (p = 0.03 and p = 0.02, respectively), decreased proportions of IgG2 (p = 0.007), and greater ratios of “upstream” to “downstream” IgG subclasses (p = 0.001) compared with HC. Serum IgG3 proportions (%IgG) from people with CIS correlated with the frequency of plasmablasts in peripheral blood (p = 0.02). Expression of Helios by Treg and Tfr cell subsets from individuals with CIS correlated with levels of serum IgG2 and IgG4. IgG3 levels and proportions of IgG3 (%IgG) in serum at CIS diagnosis were inversely correlated with the time until conversion to MS (p = 0.018 and p < 0.001, respectively), suggesting they may be useful prognostic markers of individuals with CIS who rapidly convert to MS.
Collapse
Affiliation(s)
- Stephanie Trend
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Anderson P Jones
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Lilian Cha
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Scott N Byrne
- Sydney Medical School, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Sian Geldenhuys
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Marzena J Fabis-Pedrini
- Centre for Neuromuscular and Neurological Disorders, Perron Institute for Neurological and Translational Science, Sir Charles Gairdner Hospital, University of Western Australia, Perth, WA, Australia
| | - William M Carroll
- Centre for Neuromuscular and Neurological Disorders, Perron Institute for Neurological and Translational Science, Sir Charles Gairdner Hospital, University of Western Australia, Perth, WA, Australia
| | - Judith M Cole
- St John of God Dermatology Clinic, St John of God Hospital, Perth, WA, Australia
| | - David R Booth
- Sydney Medical School, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Robyn M Lucas
- National Centre for Epidemiology & Population Health, Research School of Population Health, Australian National University, Canberra, ACT, Australia.,Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, WA, Australia
| | - Allan G Kermode
- Centre for Neuromuscular and Neurological Disorders, Perron Institute for Neurological and Translational Science, Sir Charles Gairdner Hospital, University of Western Australia, Perth, WA, Australia.,Institute for Immunology and Infectious Disease, Murdoch University, Perth, WA, Australia
| | - Martyn A French
- UWA Medical School and School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Prue H Hart
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
27
|
Pál I, Szamosi S, Hodosi K, Szekanecz Z, Váróczy L. Effect of Fcγ-receptor 3a ( FCGR3A) gene polymorphisms on rituximab therapy in Hungarian patients with rheumatoid arthritis. RMD Open 2017; 3:e000485. [PMID: 29177079 PMCID: PMC5687551 DOI: 10.1136/rmdopen-2017-000485] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 01/19/2023] Open
Abstract
Background Rheumatoid arthritis (RA) treatment includes the use of the anti-CD20 monoclonal antibody rituximab (RTX). RTX acts through Fcγ-receptors (FCGR) on effector natural killer cells and macrophages and it can be administered effectively in RA and in lymphomas. Based on the results of in vitro experiments, its efficacy may depend of FCGR gene polymorphisms in both diseases. Aim As genetic background of diseases and therapeutic efficacy (pharmacogenetics) may vary among different geographical regions, we wished to assess possible relationships between FCGR3A polymorphism and the therapeutic outcome of RTX therapy in a Hungarian RA cohort. Patients and methods Altogether, 52 patients, 6 men and 46 women, were included in the study. Peripheral blood samples were used to determine FCGR3A polymorphism by genotyping using real-time PCR method. Results The distribution of FCGR3A genotypes was 8 VV, 34 VF and 10 FF. Disease activity score 28 (DAS28) reductions in patients with VV, VF and FF genotypes were 1.98±0.54 (p=0.008 between DAS28 before and after treatment), 2.07±0.23 (p<0.001) and 1.59±0.52 (p=0.014), respectively. Significant differences in DAS28 reductions on treatment were found between VF heterozygotes and FF homozygotes (p=0.032), as well as between heterozygotes and all (VV+FF) homozygotes (p=0.017). Furthermore, significantly more VV (62.5%; p=0.030) and VF (64.7%; p=0.015) patients achieved low disease activity compared with FF subjects (30.0%). Conclusion Our results suggest that FCGR3A polymorphism may predict more effective disease activity reduction by RTX. Furthermore, carrying the V allele may also be associated with better therapeutic response in Hungarian patients with RA.
Collapse
Affiliation(s)
- Ildikó Pál
- Department of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary.,Department of Hematology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Szilvia Szamosi
- Department of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Katalin Hodosi
- Department of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary.,Department of Hematology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Zoltan Szekanecz
- Department of Rheumatology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - László Váróczy
- Department of Hematology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| |
Collapse
|
28
|
Marshall MJE, Stopforth RJ, Cragg MS. Therapeutic Antibodies: What Have We Learnt from Targeting CD20 and Where Are We Going? Front Immunol 2017; 8:1245. [PMID: 29046676 PMCID: PMC5632755 DOI: 10.3389/fimmu.2017.01245] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/19/2017] [Indexed: 12/18/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) have become one of the fastest growing classes of drugs in recent years and are approved for the treatment of a wide range of indications, from cancer to autoimmune disease. Perhaps the best studied target is the pan B-cell marker CD20. Indeed, the first mAb to receive approval by the Food and Drug Administration for use in cancer treatment was the CD20-targeting mAb rituximab (Rituxan®). Since its approval for relapsed/refractory non-Hodgkin's lymphoma in 1997, rituximab has been licensed for use in the treatment of numerous other B-cell malignancies, as well as autoimmune conditions, including rheumatoid arthritis. Despite having a significant impact on the treatment of these patients, the exact mechanisms of action of rituximab remain incompletely understood. Nevertheless, numerous second- and third-generation anti-CD20 mAbs have since been developed using various strategies to enhance specific effector functions thought to be key for efficacy. A plethora of knowledge has been gained during the development and testing of these mAbs, and this knowledge can now be applied to the design of novel mAbs directed to targets beyond CD20. As we enter the "post-rituximab" era, this review will focus on the lessons learned thus far through investigation of anti-CD20 mAb. Also discussed are current and future developments relating to enhanced effector function, such as the ability to form multimers on the target cell surface. These strategies have potential applications not only in oncology but also in the improved treatment of autoimmune disorders and infectious diseases. Finally, potential approaches to overcoming mechanisms of resistance to anti-CD20 therapy are discussed, chiefly involving the combination of anti-CD20 mAbs with various other agents to resensitize patients to treatment.
Collapse
Affiliation(s)
- Michael J. E. Marshall
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Richard J. Stopforth
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Mark S. Cragg
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
29
|
Ogura M, Coiffier B, Kwon HC, Yoon SW. Scientific rationale for extrapolation of biosimilar data across cancer indications: case study of CT-P10. Future Oncol 2017; 13:45-53. [DOI: 10.2217/fon-2017-0156] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
For a biosimilar to gain regulatory approval, a comprehensive comparability exercise must demonstrate that it is highly similar to its originator biologic, or reference product. Once biosimilarity has been shown, it is possible to approve the biosimilar for additional indications held by the reference product, without clinical trials in these indications. Extrapolation of clinical data is permitted by regulatory agencies as long as it is scientifically justified. CT-P10, a biosimilar of rituximab, was recently approved in Europe for all indications held by its reference product, incorporating both autoimmune diseases and hematological cancers. Here, we review the scientific rationale for extrapolation in biosimilar development using the example of CT-P10 as a case study.
Collapse
Affiliation(s)
- Michinori Ogura
- Department of Hematology, Tokai Central Hospital, Gifu, Japan
| | | | - Hyuk-Chan Kwon
- CELLTRION Healthcare Co. Ltd, Incheon, Republic of Korea
| | - Sang Wook Yoon
- CELLTRION Healthcare Co. Ltd, Incheon, Republic of Korea
| |
Collapse
|
30
|
Madanchi N, Bitzan M, Takano T. Rituximab in Minimal Change Disease: Mechanisms of Action and Hypotheses for Future Studies. Can J Kidney Health Dis 2017; 4:2054358117698667. [PMID: 28540057 PMCID: PMC5433659 DOI: 10.1177/2054358117698667] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/23/2016] [Indexed: 12/13/2022] Open
Abstract
Treatment with rituximab, a monoclonal antibody against the B-lymphocyte surface protein CD20, leads to the depletion of B cells. Recently, rituximab was reported to effectively prevent relapses of glucocorticoid-dependent or frequently relapsing minimal change disease (MCD). MCD is thought to be T-cell mediated; how rituximab controls MCD is not understood. In this review, we summarize key clinical studies demonstrating the efficacy of rituximab in idiopathic nephrotic syndrome, mainly MCD. We then discuss immunological features of this disease and potential mechanisms of action of rituximab in its treatment based on what is known about the therapeutic action of rituximab in other immune-mediated disorders. We believe that studies aimed at understanding the mechanisms of action of rituximab in MCD will provide a novel approach to resolve the elusive immune pathophysiology of MCD.
Collapse
Affiliation(s)
- Nima Madanchi
- Division of Nephrology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Martin Bitzan
- Division of Nephrology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Tomoko Takano
- Division of Nephrology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
31
|
Capuano C, Pighi C, Molfetta R, Paolini R, Battella S, Palmieri G, Giannini G, Belardinilli F, Santoni A, Galandrini R. Obinutuzumab-mediated high-affinity ligation of FcγRIIIA/CD16 primes NK cells for IFNγ production. Oncoimmunology 2017; 6:e1290037. [PMID: 28405525 PMCID: PMC5384385 DOI: 10.1080/2162402x.2017.1290037] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/24/2017] [Accepted: 01/26/2017] [Indexed: 01/30/2023] Open
Abstract
Natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity (ADCC), based on the recognition of IgG-opsonized targets by the low-affinity receptor for IgG FcγRIIIA/CD16, represents one of the main mechanisms by which therapeutic antibodies (mAbs) mediate their antitumor effects. Besides ADCC, CD16 ligation also results in cytokine production, in particular, NK-derived IFNγ is endowed with a well-recognized role in the shaping of adaptive immune responses. Obinutuzumab is a glycoengineered anti-CD20 mAb with a modified crystallizable fragment (Fc) domain designed to increase the affinity for CD16 and consequently the killing of mAb-opsonized targets. However, the impact of CD16 ligation in optimized affinity conditions on NK functional program is not completely understood. Herein, we demonstrate that the interaction of NK cells with obinutuzumab-opsonized cells results in enhanced IFNγ production as compared with parental non-glycoengineered mAb or the reference molecule rituximab. We observed that affinity ligation conditions strictly correlate with the ability to induce CD16 down-modulation and lysosomal targeting of receptor-associated signaling elements. Indeed, a preferential degradation of FcεRIγ chain and Syk kinase was observed upon obinutuzumab stimulation independently from CD16-V158F polymorphism. Although the downregulation of FcεRIγ/Syk module leads to the impairment of cytotoxic function induced by NKp46 and NKp30 receptors, obinutuzumab-experienced cells exhibit an increased ability to produce IFNγ in response to different stimuli. These data highlight a relationship between CD16 aggregation conditions and the ability to promote a degradative pathway of CD16-coupled signaling elements associated to the shift of NK functional program.
Collapse
Affiliation(s)
- Cristina Capuano
- Department of Experimental Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Chiara Pighi
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Simone Battella
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Gabriella Palmieri
- Department of Experimental Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Francesca Belardinilli
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Ricciarda Galandrini
- Department of Experimental Medicine, Laboratorio Pasteur Italia Fondazione Cenci Bolognetti, Sapienza University , Rome, Italy
| |
Collapse
|
32
|
Hashimoto T, Yoshida K, Hashimoto N, Nakai A, Kaneshiro K, Suzuki K, Kawasaki Y, Shibanuma N, Hashiramoto A. Circulating cell free DNA: a marker to predict the therapeutic response for biological DMARDs in rheumatoid arthritis. Int J Rheum Dis 2016; 20:722-730. [DOI: 10.1111/1756-185x.12959] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Teppei Hashimoto
- Department of Internal Medicine; Kobe University Graduate School of Medicine; Kobe Japan
- Department of Rheumatology; Kobe Kaisei Hospital; Kobe Japan
| | - Kohsuke Yoshida
- Department of Biophysics; Kobe University Graduate School of Health Sciences; Kobe Japan
| | - Naonori Hashimoto
- Department of Biophysics; Kobe University Graduate School of Health Sciences; Kobe Japan
| | - Ayako Nakai
- Department of Biophysics; Kobe University Graduate School of Health Sciences; Kobe Japan
| | - Kenta Kaneshiro
- Department of Biophysics; Kobe University Graduate School of Health Sciences; Kobe Japan
| | - Kohjin Suzuki
- Department of Biophysics; Kobe University Graduate School of Health Sciences; Kobe Japan
| | | | - Nao Shibanuma
- Department of Rheumatology; Kobe Kaisei Hospital; Kobe Japan
- Department of Orthopedic Surgery; Kobe Kaisei Hospital; Kobe Japan
| | - Akira Hashiramoto
- Department of Biophysics; Kobe University Graduate School of Health Sciences; Kobe Japan
| |
Collapse
|
33
|
Anaya JM, Duarte-Rey C, Sarmiento-Monroy JC, Bardey D, Castiblanco J, Rojas-Villarraga A. Personalized medicine. Closing the gap between knowledge and clinical practice. Autoimmun Rev 2016; 15:833-42. [DOI: 10.1016/j.autrev.2016.06.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 06/07/2016] [Indexed: 12/22/2022]
|
34
|
Awdishu L, Joy MS. Role of Pharmacogenomics in Kidney Disease and Injury. Adv Chronic Kidney Dis 2016; 23:106-19. [PMID: 26979149 DOI: 10.1053/j.ackd.2016.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/28/2016] [Accepted: 01/31/2016] [Indexed: 12/29/2022]
Abstract
There has been considerable excitement in the kidney community surrounding the research findings on the genetic contributions to kidney diseases. However, positive outcomes of personalized therapeutic interventions can be circumvented by unpredictable pharmacokinetics of prescribed drugs. Furthermore, unpredictable drug disposition can result in toxicities such as kidney injury. Patient covariates, disease covariates, and pharmacogenetics all contribute to variability in drug disposition. Further treatment personalization and avoidance of drug- and biologic- induced kidney injury will require extensive knowledge and expertise in renal clinical pharmacology. The current review will focus on the pharmacogenetics of drugs and biologics used in the treatment of glomerular kidney diseases and drugs implicated in inducing kidney injury phenotypes.
Collapse
|
35
|
Montes A, Perez-Pampin E, Joven B, Carreira P, Fernández-Nebro A, Del Carmen Ordóñez M, Navarro-Sarabia F, Moreira V, Vasilopoulos Y, Sarafidou T, Caliz R, Ferrer MA, Cañete JD, de la Serna AR, Magallares B, Narváez J, Gómez-Reino JJ, Gonzalez A. FCGR polymorphisms in the treatment of rheumatoid arthritis with Fc-containing TNF inhibitors. Pharmacogenomics 2016; 16:333-45. [PMID: 25823782 DOI: 10.2217/pgs.14.175] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES Reproducible association of a functional polymorphism in FCGR2A with response to a TNF inhibitor (TNFi) in patients with rheumatoid arthritis (RA) led us to explore other FcγR functional polymorphisms. METHODS Functional polymorphisms FCGR3A F158V, FCGR2B I223T and promoter VNTR in FCGRT were analyzed in up to 429 patients with RA. Response to TNFi was recorded during standard care at 3, 6 and 12 months of follow-up. Fixed effects meta-analysis of studies addressing FCGR3A F158V polymorphism, which is the most studied of these polymorphisms, was conducted with inverse variance weighting. RESULTS None of the functional polymorphisms were associated with change in DAS28. Meta-analysis of the seven studies (899 patients) with available data addressing association of FCGR3A F158V with response to TNFi in RA showed no association (OR: 1.11, 95% CI: 0.8-1.5; p = 0.5). CONCLUSION None of the three functional polymorphisms in FcγR genes showed association with response to TNFi in patients with RA. These negative results were obtained in spite of the larger size of this study relative to previous studies addressing the same polymorphisms. In addition, meta-analysis of FCGR3A F158V was also negative against the results provided by previous studies. Original submitted 17 September 2014; Revision submitted 9 December 2014.
Collapse
Affiliation(s)
- Ariana Montes
- Laboratorio de Investigacion 10 & Rheumatology Unit, Instituto de Investigacion Sanitaria - Hospital Clinico Universitario de Santiago, Travesia da Choupana s/n, 15706, Santiago de Compostela, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Cuppen BVJ, Welsing PMJ, Sprengers JJ, Bijlsma JWJ, Marijnissen ACA, van Laar JM, Lafeber FPJG, Nair SC. Personalized biological treatment for rheumatoid arthritis: a systematic review with a focus on clinical applicability. Rheumatology (Oxford) 2015; 55:826-39. [PMID: 26715775 DOI: 10.1093/rheumatology/kev421] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 01/30/2023] Open
Abstract
OBJECTIVES To review studies that address prediction of response to biologic treatment in RA and to explore the clinical utility of the studied (bio)markers. METHODS A search for relevant articles was performed in PubMed, Embase and Cochrane databases. Studies that presented predictive values or in which these could be calculated were selected. The added value was determined by the added value on prior probability for each (bio)marker. Only an increase/decrease in chance of response ⩾15% was considered clinically relevant, whereas in oncology values >25% are common. RESULTS Of the 57 eligible studies, 14 (bio)markers were studied in more than one cohort and an overview of the added predictive value of each marker is presented. Of the replicated predictors, none consistently showed an increase/decrease in probability of response ⩾15%. However, positivity of RF and ACPA in case of rituximab and the presence of the TNF-α promoter 308 GG genotype for TNF inhibitor therapy were consistently predictive, yet low in added predictive value. Besides these, 65 (bio)markers studied once showed remarkably high (but not validated) predictive values. CONCLUSION We were unable to address clinically useful baseline (bio)markers for use in individually tailored treatment. Some predictors are consistently predictive, yet low in added predictive value, while several others are promising but await replication. The challenge now is to design studies to validate all explored and promising findings individually and in combination to make these (bio)markers relevant to clinical practice.
Collapse
Affiliation(s)
- Bart V J Cuppen
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paco M J Welsing
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan J Sprengers
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johannes W J Bijlsma
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anne C A Marijnissen
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jacob M van Laar
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Floris P J G Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sandhya C Nair
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
37
|
Tarnowski M, Paradowska-Gorycka A, Dąbrowska-Zamojcin E, Czerewaty M, Słuczanowska-Głąbowska S, Pawlik A. The effect of gene polymorphisms on patient responses to rheumatoid arthritis therapy. Expert Opin Drug Metab Toxicol 2015; 12:41-55. [PMID: 26609565 DOI: 10.1517/17425255.2016.1121233] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a systemic disease leading to joint destruction. The therapy of RA is mainly based on disease-modifying anti-rheumatic drugs (DMARDs) and biological drugs. The response to treatment is different among patients. Therefore, we have searched for factors that may predict the efficacy and toxicity during therapy in individual patients. AREAS COVERED This review presents the role of genetic polymorphisms as predictors of the efficacy and toxicity during the therapy of RA patients with DMARDs (methotrexate, leflunomide, sulfasalazine) and biological drugs (anti-TNF-alpha antagonists, Tocilizumab, Rituximab). EXPERT OPINION Despite studies having shown an association between genetic polymorphisms and response to therapy in RA patients, the majority of these findings are still inconclusive and inconsistent. We are still far from applying pharmacogenetic tests in routine clinical practice that can predict the outcome of treatment. Several factors, such as small sample size with low statistical power, variability in the outcome definitions and the heterogeneity of the cohorts, limited number of tested single nucleotide polymorphisms (SNPs), small effect for the selected variant, and a lack of consideration of epigenetic factors, may contribute to the inconsistency observed and may lead to limited success in personalizing therapy.
Collapse
Affiliation(s)
- Maciej Tarnowski
- a Department of Physiology , Pomeranian Medical University , 70-111 Szczecin , Poland
| | - Agnieszka Paradowska-Gorycka
- b Department of Biochemistry and Molecular Biology , National Institute of Geriatrics, Rheumatology and Rehabilitation , 02-637 Warszawa , Poland
| | | | - Michal Czerewaty
- a Department of Physiology , Pomeranian Medical University , 70-111 Szczecin , Poland
| | | | - Andrzej Pawlik
- a Department of Physiology , Pomeranian Medical University , 70-111 Szczecin , Poland
| |
Collapse
|
38
|
Abstract
BACKGROUND Treatment with rituximab may be accompanied by a systemic cytokine release. We studied the effects of a single dose of rituximab on cytokine levels in transplant patients and examined the underlying mechanism. METHODS Twenty renal transplant recipients (10 rituximab-treated, 10 placebo-treated) were recruited from a randomized clinical trial. Rituximab or placebo was infused during surgery, and blood samples were taken before, during, and after surgery and analyzed for interleukin (IL)-2, IL-4, IL-6, IL-10, IL-12, IL-17, interferon-γ, macrophage inflammatory protein (MIP)-1β, transforming growth factor-β, and tumor necrosis factor-α. in vitro, healthy donor peripheral blood mononuclear cells, purified B cells, monocytes, natural killer (NK) cells, or combinations thereof were incubated with rituximab, rituximab-F(ab')2, or medium and MIP-1β, IL-10, interferon-γ, and tumor necrosis factor-α levels were measured in the supernatant. RESULTS Rituximab-treated patients had higher serum levels of IL-10 (101 ± 35 pg/mL vs 41 ± 9 pg/mL; P < 0.01) and MIP-1β (950 ± 418 pg/mL vs 125 ± 32 pg/mL; P < 0.001) compared to placebo-treated patients at 2 hours after start of infusion. There was no difference in the level of other cytokines. In vitro, the addition of rituximab, but not rituximab-F(ab')2 fragments, only led to significantly increased levels of MIP-1β in co-cultures of B and NK cells. Levels of MIP-1β were higher in patients with a high affinity Fc-receptor compared to those with a lower affinity FcγRIIIa (1356 ± 184 pg/mL vs 679 ± 273 pg/mL; P < 0.01). CONCLUSIONS In addition to B-cell depletion, rituximab can modulate the immune response by inducing cytokine secretion, especially IL-10 and MIP-1β. Rituximab-induced MIP-1β secretion depends on the combined presence of B cells and FcR-bearing cells, especially NK cells.
Collapse
|
39
|
Hargreaves CE, Rose-Zerilli MJJ, Machado LR, Iriyama C, Hollox EJ, Cragg MS, Strefford JC. Fcγ receptors: genetic variation, function, and disease. Immunol Rev 2015; 268:6-24. [DOI: 10.1111/imr.12341] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Chantal E. Hargreaves
- Cancer Genomics Group; Cancer Sciences; Faculty of Medicine; University of Southampton; Southampton UK
- Antibody and Vaccine Group; Cancer Sciences; Faculty of Medicine; University of Southampton; Southampton UK
| | | | - Lee R. Machado
- Department of Genetics; University of Leicester; Leicester UK
- School of Health; University of Northampton; Northampton UK
| | - Chisako Iriyama
- Department of Hematology and Oncology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | | | - Mark S. Cragg
- Antibody and Vaccine Group; Cancer Sciences; Faculty of Medicine; University of Southampton; Southampton UK
| | - Jonathan C. Strefford
- Cancer Genomics Group; Cancer Sciences; Faculty of Medicine; University of Southampton; Southampton UK
| |
Collapse
|
40
|
Reddy V, Cambridge G, Isenberg DA, Glennie MJ, Cragg MS, Leandro M. Internalization of rituximab and the efficiency of B Cell depletion in rheumatoid arthritis and systemic lupus erythematosus. Arthritis Rheumatol 2015; 67:2046-55. [PMID: 25916583 PMCID: PMC4737120 DOI: 10.1002/art.39167] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 04/16/2015] [Indexed: 12/26/2022]
Abstract
Objective Rituximab, a type I anti‐CD20 monoclonal antibody (mAb), induces incomplete B cell depletion in some patients with rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), thus contributing to a poor clinical response. The mechanisms of this resistance remain elusive. The purpose of this study was to determine whether type II mAb are more efficient than type I mAb at depleting B cells from RA and SLE patients, whether internalization influences the efficiency of depletion, and whether Fcγ receptor type IIb (FcγRIIb) and the B cell receptor regulate this internalization process. Methods We used an in vitro whole blood B cell–depletion assay to assess the efficiency of depletion, flow cytometry to study cell surface protein expression, and surface fluorescence–quenching assays to assess rituximab internalization, in samples from patients with RA and patients with SLE. Paired t‐test or Mann‐Whitney U test was used to compare groups, and Spearman's rank correlation test was used to assess correlation. Results We found that type II mAb internalized significantly less rituximab than type I mAb and depleted B cells from patients with RA and SLE at least 2‐fold more efficiently than type I mAb. Internalization of rituximab was highly variable between patients, was regulated by FcγRIIb, and inversely correlated with cytotoxicity in whole blood B cell–depletion assays. The lowest levels of internalization were seen in IgD– B cells, including postswitched (IgD–CD27+) memory cells. Internalization of type I anti‐CD20 mAb was also partially inhibited by anti‐IgM stimulation. Conclusion Variability in internalization of rituximab was observed and was correlated with impaired B cell depletion. Therefore, slower‐internalizing type II mAb should be considered as alternative B cell–depleting agents for the treatment of RA and SLE.
Collapse
|
41
|
Anders HJ, Weidenbusch M, Rovin B. Unmet medical needs in lupus nephritis: solutions through evidence-based, personalized medicine. Clin Kidney J 2015; 8:492-502. [PMID: 26413272 PMCID: PMC4581390 DOI: 10.1093/ckj/sfv072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/17/2015] [Indexed: 12/12/2022] Open
Abstract
Lupus nephritis (LN) remains a kidney disease with significant unmet medical needs despite extensive clinical and translational research over the past decade. These include the need to (i) predict the individual risk for LN in a patient with systemic lupus erythematosus, (ii) identify the best therapeutic option for an individual patient, (iii) distinguish chronic kidney damage from active immunologic kidney injury, (iv) develop efficient treatments with acceptable or no side effects and improve the design of randomized clinical trials so that effective drugs demonstrate efficacy. This review discusses the underlying reasons for these unmet medical needs and options of how to overcome them in the future.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Marc Weidenbusch
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Brad Rovin
- Division of Nephrology, Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
42
|
Accurate interrogation of FCGR3A rs396991 in European and Asian populations using a widely available TaqMan genotyping method. Pharmacogenet Genomics 2015; 25:569-72. [PMID: 26367501 PMCID: PMC4596484 DOI: 10.1097/fpc.0000000000000175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Supplemental Digital Content is available in the text. A polymorphism in the receptor for the Fc region of IgG, Fc γ-receptor IIIa (FcγRIIIa, FCGR3A rs396991), has been inconsistently shown in the literature to have an effect on response to monoclonal antibody therapy in several indications. The rs396991 (T/G) polymorphism leads to an F176V substitution and increased affinity for IgG. This variant has proven difficult to genotype accurately, primarily because of extensive homology between the FCGR3A and FCGR3B genes. We have shown that rs396991 can be genotyped by PCR amplification, followed by direct Sanger sequencing of the product, without coamplification of FCGR3B, and that the rs396991 TaqMan assay (C__25815666_10) agrees with Sanger sequencing results in 100% of European and Asian samples tested, but it has a small error rate in African and American populations. C__25815666_10 is therefore suitable to interrogate rs396991 in studies involving Europeans and Asians; however for other populations, the default genotyping method should be PCR followed by Sanger sequencing.
Collapse
|
43
|
Fabris M, Quartuccio L, Fabro C, Sacco S, Lombardi S, Ramonda R, Biasi D, Punzi D, Adami S, Olivieri I, Curcio F, De Vita S. The -308 TNFα and the -174 IL-6 promoter polymorphisms associate with effective anti-TNFα treatment in seronegative spondyloarthritis. THE PHARMACOGENOMICS JOURNAL 2015; 16:238-42. [PMID: 26149736 DOI: 10.1038/tpj.2015.49] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 04/07/2015] [Accepted: 05/21/2015] [Indexed: 12/27/2022]
Abstract
The genetic predisposition to a long-term efficacy of anti-tumor necrosis factor (TNF)α treatment in seronegative spondyloarthritis (SpA) was investigated by analysing the possible correlation between several single nucleotide gene polymorphisms and the retention rate of anti-TNFα therapies. We compared patients needing to switch the first anti-TNFα (Sw, No. 64) within at least 12 months of follow-up with patients not needing to switch (NSw, No. 123), observing at least 6 months of treatment to establish anti-TNFα failure, leading to treatment change. Response to treatment was evaluated by standardised criteria (BASDAI for axial involvement, DAS28-EULAR for peripheral involvement). The TNFα -308 A allele and the interleukin (IL)-6 -174GG homozygosis resulted as independent biomarkers predicting survival of the first anti-TNFα therapy in SpA patients (P=0.007, odds ratio (OR): 4.4, 95% confidence interval (CI)=1.5-13.1 and P=0.035, OR: 2.1, 95% CI=1.1-4.4). Also, the male gender (P=0.001, OR: 3.4, 95% CI=1.6-7.1) associated with the NSw phenotype, whereas no association was found either with the specific diagnosis or the predominant joint involvement.
Collapse
Affiliation(s)
- M Fabris
- Clinical Pathology, University-Hospital of Udine, Udine, Italy.,Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - L Quartuccio
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy.,Rheumatology, University-Hospital of Udine, Udine, Italy
| | - C Fabro
- Rheumatology, University-Hospital of Udine, Udine, Italy
| | - S Sacco
- Rheumatology, University-Hospital of Udine, Udine, Italy
| | - S Lombardi
- Institute of Physical Medicine and Rehabilitation 'Gervasutta', Udine, Italy
| | - R Ramonda
- Rheumatology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - D Biasi
- Section of Rheumatology, Department of Medicine, University of Verona, Verona, Italy
| | - D Punzi
- Rheumatology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - S Adami
- Section of Rheumatology, Department of Medicine, University of Verona, Verona, Italy
| | - I Olivieri
- Rheumatology Department of Lucania, San Carlo Hospital of Potenza, Potenza, Italy
| | - F Curcio
- Clinical Pathology, University-Hospital of Udine, Udine, Italy.,Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | - S De Vita
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy.,Rheumatology, University-Hospital of Udine, Udine, Italy
| |
Collapse
|
44
|
Liu L. Antibody Glycosylation and Its Impact on the Pharmacokinetics and Pharmacodynamics of Monoclonal Antibodies and Fc-Fusion Proteins. J Pharm Sci 2015; 104:1866-1884. [DOI: 10.1002/jps.24444] [Citation(s) in RCA: 246] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/27/2015] [Accepted: 03/17/2015] [Indexed: 12/12/2022]
|
45
|
Gibson DS, Bustard MJ, McGeough CM, Murray HA, Crockard MA, McDowell A, Blayney JK, Gardiner PV, Bjourson AJ. Current and future trends in biomarker discovery and development of companion diagnostics for arthritis. Expert Rev Mol Diagn 2014; 15:219-34. [PMID: 25455156 DOI: 10.1586/14737159.2015.969244] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Musculoskeletal diseases such as rheumatoid arthritis are complex multifactorial disorders that are chronic in nature and debilitating for patients. A number of drug families are available to clinicians to manage these disorders but few tests exist to target these to the most responsive patients. As a consequence, drug failure and switching to drugs with alternate modes of action is common. In parallel, a limited number of laboratory tests are available which measure biological indicators or 'biomarkers' of disease activity, autoimmune status, or joint damage. There is a growing awareness that assimilating the fields of drug selection and diagnostic tests into 'companion diagnostics' could greatly advance disease management and improve outcomes for patients. This review aims to highlight: the current applications of biomarkers in rheumatology with particular focus on companion diagnostics; developments in the fields of proteomics, genomics, microbiomics, imaging and bioinformatics and how integration of these technologies into clinical practice could support therapeutic decisions.
Collapse
Affiliation(s)
- David S Gibson
- Northern Ireland Centre for Stratified Medicine, University of Ulster, C-TRIC Building, Altnagelvin Hospital campus, Glenshane Road, Londonderry, BT47 6SB, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bonatti F, Reina M, Neri TM, Martorana D. Genetic Susceptibility to ANCA-Associated Vasculitis: State of the Art. Front Immunol 2014; 5:577. [PMID: 25452756 PMCID: PMC4233908 DOI: 10.3389/fimmu.2014.00577] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 10/28/2014] [Indexed: 12/12/2022] Open
Abstract
ANCA-associated vasculitis (AAV) is a group of disorders that is caused by inflammation affecting small blood vessels. Both arteries and veins are affected. AAV includes microscopic polyangiitis (MPA), granulomatosis with polyangiitis (GPA) renamed from Wegener’s granulomatosis, and eosinophilic granulomatosis with polyangiitis (EGPA), renamed from Churg–Strauss syndrome. AAV is primarily due to leukocyte migration and resultant damage. Despite decades of research, the mechanisms behind AAV disease etiology are still not fully understood, although it is clear that genetic and environmental factors are involved. To improve the understanding of the disease, the genetic component has been extensively studied by candidate association studies and two genome-wide association studies. The majority of the identified genetic AAV risk factors are common variants. These have uncovered information that still needs further investigation to clarify its importance. In this review, we summarize and discuss the results of the genetic studies in AAV. We also present the novel approaches to identifying the causal variants in complex susceptibility loci and disease mechanisms. Finally, we discuss the limitations of current methods and the challenges that we still have to face in order to incorporate genomic and epigenomic data into clinical practice.
Collapse
Affiliation(s)
- Francesco Bonatti
- Unit of Medical Genetics, Laboratory of Molecular Genetics, Diagnostic Department, University Hospital of Parma , Parma , Italy
| | - Michele Reina
- Unit of Medical Genetics, Laboratory of Molecular Genetics, Diagnostic Department, University Hospital of Parma , Parma , Italy
| | - Tauro Maria Neri
- Unit of Medical Genetics, Laboratory of Molecular Genetics, Diagnostic Department, University Hospital of Parma , Parma , Italy
| | - Davide Martorana
- Unit of Medical Genetics, Laboratory of Molecular Genetics, Diagnostic Department, University Hospital of Parma , Parma , Italy
| |
Collapse
|
47
|
Therapeutic options after treatment failure in rheumatoid arthritis or spondyloarthritides. Adv Ther 2014; 31:780-802. [PMID: 25112460 DOI: 10.1007/s12325-014-0142-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Indexed: 02/08/2023]
Abstract
The prognosis for patients with rheumatoid arthritis or spondyloarthritides has improved dramatically due to earlier diagnosis, recognition of the need to treat early with conventional synthetic disease-modifying antirheumatic drugs (csDMARDs), alone or in combinations, the establishment of treatment targets, and the development of biological DMARDs (bDMARDs). Many patients are now able to achieve clinical remission or low disease activity with therapy, and reduce or eliminate systemic corticosteroid use. Guidelines recommend methotrexate as a first-line agent for the initial treatment of rheumatoid arthritis; however, a majority of patients will require a change of csDMARD or step up to combination therapy with the addition of another csDMARD or a bDMARD. However, treatment failure is common and switching to a different therapy may be required. The large number of available treatment options, combined with a lack of comparative data, makes the choice of a new therapy complex and often not evidence based. We summarize and discuss evidence to inform treatment decisions in patients who require a change in therapy, including baseline factors that may predict response to therapy.
Collapse
|
48
|
Lee YH, Bae SC, Song GG. Functional FCGR3A 158 V/F and IL-6 -174 C/G polymorphisms predict response to biologic therapy in patients with rheumatoid arthritis: a meta-analysis. Rheumatol Int 2014; 34:1409-15. [PMID: 24728031 DOI: 10.1007/s00296-014-3015-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 03/31/2014] [Indexed: 01/10/2023]
Abstract
The aim of this study was to investigate whether the Fc gamma receptor 3A (FCGR3A) 158 V/F and interleukin-6 (IL-6) promoter -174 G/C polymorphisms can predict the response to biologic-based therapy in patients with rheumatoid arthritis (RA). We conducted a meta-analysis of studies on the association between the FCGR3A V/F polymorphism or the IL-6 -174 C/G polymorphism and non-responsiveness to biologic therapy in RA patients. A total of 10 studies involving 1,427 patients were considered. These studies consisted of seven studies on the FCGR3A polymorphism and three studies on the IL-6 polymorphism. Meta-analysis showed no association between the FCGR3A VV+VF genotype and non-responders to biologic therapy [odds ratio (OR) 0.881, 95 % confidence interval (CI) 0.505-1.537, p = 0.655]. However, stratification by biologic type indicated an association between the FCGR3A VV+VF genotype and non-responders to rituximab (OR 0.566, 95 % CI 0.373-0.857, p = 0.007), but no association was found in non-responders to tumor necrosis factor (TNF)-blockers (OR 1.337, 95 % CI 0.869-2.056, p = 0.186). Meta-analysis revealed no association between the IL-6 CC+CG genotype and non-responders to the biologics (OR 3.233, 95 % CI 0.766-13.64, p = 0.110). However, an association was found between the IL-6 CC+CG genotype and non-responders to anti-TNF therapy (OR 8.030, 95 % CI 1.807-33.68, p = 0.006). This meta-analysis demonstrates that FCGR3A V allele carriers show a better response to rituximab, and individuals carrying the IL-6 -174 C allele show a poorer response to anti-TNF therapy for RA. Genotyping for these polymorphisms may be a useful tool for predicting the response to biologics with respect to personalized medicine.
Collapse
Affiliation(s)
- Young Ho Lee
- Division of Rheumatology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Korea University Medical Center, 126-1 ga, Anam-dong, Seongbuk-gu, Seoul, 136-705, Korea,
| | | | | |
Collapse
|
49
|
Attenuation of Collagen-Induced Arthritis in rat by nicotinic alpha7 receptor partial agonist GTS-21. BIOMED RESEARCH INTERNATIONAL 2014; 2014:325875. [PMID: 24719855 PMCID: PMC3955649 DOI: 10.1155/2014/325875] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/10/2014] [Indexed: 01/01/2023]
Abstract
This research was performed to observe the effect of GTS-21 on Collagen Induced Arthritis (CIA). CIA model was used and after the onset of arthritis, the rats were divided into three groups based on their clinical symptoms score. Two groups were intraperitoneally (IP) injected daily with GTS-21 (1 mg/kg, 2.5 mg/kg) for a week, whereas phosphate buffered saline (PBS) was used for the control group. Cytokine titers, radiological, and histological examinations were performed at different time points after treatment with GTS-21. Compared with those of the control, the levels of TNF-α, IL-1, and IL-6 in the serum were significantly reduced after GTS-21 management. In addition, radiological results show that bone degradation was inhibited as well. Moreover, the hematoxylin and eosin (H&E) staining indicated that the histological score was significantly alleviated in the therapeutic group. Tartrate-resistant acid phosphatase (TRAP) stain-positive cells were also detected in the destruction of the articular cartilage, which was significantly reduced compared with the control group. This study provides the first evidence on the effect of GTS-21 as a potential treatment for RA.
Collapse
|
50
|
Predictive factors of response to biological disease modifying antirheumatic drugs: towards personalized medicine. Mediators Inflamm 2014; 2014:386148. [PMID: 24523570 PMCID: PMC3913459 DOI: 10.1155/2014/386148] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 11/27/2013] [Indexed: 01/19/2023] Open
Abstract
Many therapies are now available for patients with rheumatoid arthritis (RA) who have an inadequate response to methotrexate including tumor necrosis factor inhibitors, abatacept, tocilizumab, and rituximab. Clinical response to drugs varies widely between individuals. A part of this variability is due to the characteristics of the patient such as age, gender, concomitant therapies, body mass index, or smoking status. Clinical response also depends on disease characteristics including disease activity and severity and presence of autoantibodies. Genetic background, cytokine levels, and immune cell phenotypes could also influence biological therapy response. This review summarizes the impact of all those parameters on response to biological therapies.
Collapse
|