51
|
Rhupus: a systematic literature review. Autoimmun Rev 2020; 19:102612. [DOI: 10.1016/j.autrev.2020.102612] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 01/12/2023]
|
52
|
Basta F, Fasola F, Triantafyllias K, Schwarting A. Systemic Lupus Erythematosus (SLE) Therapy: The Old and the New. Rheumatol Ther 2020; 7:433-446. [PMID: 32488652 PMCID: PMC7410873 DOI: 10.1007/s40744-020-00212-9] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Indexed: 12/18/2022] Open
Abstract
Despite recent improvements in the treatment of systemic lupus erythematosus (SLE), disease activity, comorbidities and drug toxicity significantly contribute to the risk of progressive irreversible damage accrual and increased mortality in patients with this chronic disease. Moreover, even lupus patients in remission often report residual symptoms, such as fatigue, which have a considerable impact on their health-related quality of life. In recent decades, SLE treatment has moved from the use of hydroxychloroquine, systemic glucocorticosteroids and conventional immunosuppressive drugs to biologic agents, of which belimumab is the first and only biologic agent approved for the treatment for SLE to date. Novel therapies targeting interferons, cytokines and their receptors, intracellular signals, plasma cells, T lymphocytes and co-stimulatory molecules are being evaluated. In the context of a holistic approach, growing evidence is emerging of the importance of correct lifestyle habits in the management of lupus manifestations and comorbidities. The aim of this paper is to provide an overview of current pharmacological and non-pharmacological treatment options and emerging therapies in SLE.
Collapse
Affiliation(s)
- Fabio Basta
- Acura Rheumatology Center Rhineland Palatinate, Bad Kreuznach, Germany.
- University Center of Autoimmunity, Johannes Gutenberg University, Mainz, Germany.
| | - Federica Fasola
- Acura Rheumatology Center Rhineland Palatinate, Bad Kreuznach, Germany
- University Center of Autoimmunity, Johannes Gutenberg University, Mainz, Germany
| | - Konstantinos Triantafyllias
- Acura Rheumatology Center Rhineland Palatinate, Bad Kreuznach, Germany
- University Center of Autoimmunity, Johannes Gutenberg University, Mainz, Germany
| | - Andreas Schwarting
- Acura Rheumatology Center Rhineland Palatinate, Bad Kreuznach, Germany
- Division of Rheumatology and Clinical Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
- University Center of Autoimmunity, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
53
|
Moore E, Putterman C. Are lupus animal models useful for understanding and developing new therapies for human SLE? J Autoimmun 2020; 112:102490. [PMID: 32535128 PMCID: PMC7384952 DOI: 10.1016/j.jaut.2020.102490] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 01/13/2023]
Abstract
Systemic lupus erythematosus is a systemic autoimmune disease driven by a complex combination of genetic, environmental, and other immunoregulatory factors. The development of targeted therapies is complicated by heterogeneous clinical manifestations, varying organ involvement, and toxicity. Despite advances in understanding the mechanisms contributing to SLE, only one biologic drug, belimumab, is FDA-approved. The identification and development of potential therapies have largely been driven by studies in lupus animal models. Therefore, direct comparison of both the therapeutic and immunological findings in human and murine SLE studies is critical and can reveal important insights into indeed how useful and relevant are murine studies in SLE drug development. Studies involving belimumab, mycophenolate mofetil, abatacept, rituximab, and anti-interferon strategies generally demonstrated analogous findings in the attenuation of SLE manifestations and modulation of select immune cell populations in human and murine SLE. While further basic and translational studies are needed to identify SLE patient subsets likely to respond to particular therapeutic modalities and in dissecting complex mechanisms, we believe that despite some inherent weaknesses SLE mouse models will continue to be integral in developing targeted SLE therapies.
Collapse
Affiliation(s)
- Erica Moore
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chaim Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA; Bar-Ilan University Azrieli Faculty of Medicine, Safed, Israel; Research Institute, Galilee Medical Center, Nahariya, Israel.
| |
Collapse
|
54
|
Zhang PF, Xie D, Li Q. Chimeric antigen receptor T-cell therapy beyond cancer: current practice and future prospects. Immunotherapy 2020; 12:1021-1034. [PMID: 32727249 DOI: 10.2217/imt-2020-0009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Adoptive cell therapy with chimeric antigen receptor T (CAR-T) cells has achieved remarkable efficacy in the treatment of hematological malignancies, which has inspired researchers to expand the application of CAR-T-cell therapy to other medical conditions. Here, we review the current understanding and development of CAR-T-cell therapy for infectious diseases, autoimmune diseases and allotransplantation. The limitations and challenges of CAR-T-cell therapy in the treatment of these diseases and potential solutions to overcome these shortcomings are also discussed. With the development of novel designs of CARs and preclinical/clinical investigations, CAR-T-cell therapy is expected to be a potential cure option in a wide array of disease settings in the future.
Collapse
Affiliation(s)
- Peng-Fei Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Dan Xie
- Prenatal Diagnosis Center, Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China, 610041.,Key Laboratory of Birth Defects & Related Diseases of Women & Children (Sichuan University), Ministry of Education, Chengdu, China, 610041
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China, 610041
| |
Collapse
|
55
|
Mihaylova N, Bradyanova S, Chipinski P, Chausheva S, Kyurkchiev D, Tchorbanov AI. Monoclonal antibody therapy that targets phospholipid-binding protein delays lupus activity in MRL/lpr mice. Scand J Immunol 2020; 92:e12915. [PMID: 32533866 DOI: 10.1111/sji.12915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/14/2020] [Accepted: 06/03/2020] [Indexed: 11/28/2022]
Abstract
Systemic lupus erythematosus is an autoimmune syndrome characterized by the development of autoantibodies to a wide range of antigens. Together with B cells, respective self-reactive T cells have an important contribution in disease progression as being responsible for inflammatory cytokines secretion, B cell activation and promoting amplification of the autoimmune response. Annexin A1 is expressed by many cell types and binds to phospholipids in a Ca2+ -dependent manner. Abnormal expression of annexin A1 was found on activated B and T cells in both murine and human autoimmunity suggesting its potential role as a therapeutic target. In the present study, we have investigated the possibility to suppress autoimmune manifestation in spontaneous mouse model of lupus using anti-annexin A1 antibody. Groups of lupus-prone MRL/lpr mice were treated with the anti-annexin A1 monoclonal antibody, and the disease activity and survival of the animals were following up. Flow cytometry, ELISA assays, and histological and immunofluorescence kidney analyses were used to determine the levels of Annexin A1 expression, cytokines, anti-dsDNA antibodies and kidney injuries. The administration of this monoclonal antibody to MRL/lpr mice resulted in suppression of IgG anti-dsDNA antibody production, modulated IL-10 secretion, decreased disease activity and prolonged survival compared with the control group.
Collapse
Affiliation(s)
- Nikolina Mihaylova
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Silviya Bradyanova
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Petroslav Chipinski
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Stela Chausheva
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Dobroslav Kyurkchiev
- Laboratory of Clinical Immunology, University Hospital 'Sv.I.Rilski', Medical University Sofia, Sofia, Bulgaria
| | - Andrey I Tchorbanov
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.,National Institute of Immunology, Sofia, Bulgaria
| |
Collapse
|
56
|
Abstract
Following the advent of molecular targeted drugs, a paradigm shift in treatment similar to that in rheumatoid arthritis has been expected in the treatment of systemic lupus erythematosus (SLE), but clinical trials for drugs that many specialists believed to be effective have failed repeatedly. The causes are not simple, but include the heterogeneity of SLE, inclusion criteria, lack of appropriate disease activity measures, and relapse criteria. This review outlines the disease activity indices used in SLE, discusses their advantages and disadvantages, and describes the ideal activity index.
Collapse
Affiliation(s)
- Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
57
|
Hoyer BF. Neue Therapien beim systemischen Lupus erythematodes. Z Rheumatol 2020; 79:342-350. [DOI: 10.1007/s00393-020-00788-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
58
|
|
59
|
Krummey SM, Hartigan CR, Liu D, Ford ML. CD28-Dependent CTLA-4 Expression Fine-Tunes the Activation of Human Th17 Cells. iScience 2020; 23:100912. [PMID: 32203908 PMCID: PMC7096747 DOI: 10.1016/j.isci.2020.100912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 09/26/2019] [Accepted: 02/10/2020] [Indexed: 01/05/2023] Open
Abstract
Previous work has demonstrated that Th17 memory cells but not Th1 cells are resistant to CD28/CTLA-4 blockade with CTLA-4 Ig, leading us to investigate the individual roles of the CD28 and CTLA-4 cosignaling pathways on Th1 versus Th17 cells. We found that selective CD28 blockade with a domain antibody (dAb) inhibited Th1 cells but surprisingly augmented Th17 responses. CD28 agonism resulted in a profound increase in CTLA-4 expression in Th17 cells as compared with Th1 cells. Consistent with these findings, inhibition of the CD28 signaling protein AKT revealed that CTLA-4 expression on Th17 cells was more significantly reduced by AKT inhibition relative to CTLA-4 expression on Th17 cells. Finally, we found that FOXO1 and FOXO3 overexpression restrained high expression of CTLA-4 on Th17 cells but not Th1 cells. This study demonstrates that the heterogeneity of the CD4+ T cell compartment has implications for the immunomodulation of pathologic T cell responses. CD28 blockade resulted in augmentation of human Th17 cells relative to Th1 cells Th17 polarized mice exhibited graft rejection in the presence of CD28 blockade A significant portion of Th17 cell CTLA-4 expression was induced by CD28 ligation Overexpression of FOXO1 or FOXO3 inhibited Th17 cell CTLA-4 expression
Collapse
Affiliation(s)
- Scott M Krummey
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Christina R Hartigan
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Danya Liu
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Mandy L Ford
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
60
|
Tsang-A-Sjoe MWP, Bultink IEM, Heslinga M, van Tuyl LH, van Vollenhoven RF, Voskuyl AE. The relationship between remission and health-related quality of life in a cohort of SLE patients. Rheumatology (Oxford) 2020; 58:628-635. [PMID: 30517706 DOI: 10.1093/rheumatology/key349] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/18/2018] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To investigate the relationship between remission and health-related quality of life (HRQoL) in patients with SLE in a longitudinal observational cohort. METHODS HRQoL was measured at cohort visits using the physical and mental component score (PCS and MCS, respectively) of the Short Form 36 questionnaire. Definitions of Remission in SLE remission categories (no remission/remission on therapy/remission off therapy) were applied. Determinants of PCS and MCS were identified with simple linear regression analyses. Association between remission and HRQoL was assessed using generalized estimating equation models. RESULTS Data from 154 patients with 2 years of follow-up were analysed. At baseline 60/154 (39.0%) patients were in either form of remission. Patients in remission had higher Short Form 36 scores in all subdomains compared with patients not in remission. PCS was positively associated with remission and employment, and negatively associated with SLICC damage index, ESR, medication, patient global assessment and BMI. MCS was positively associated with Caucasian ethnicity and negatively associated with patient global assessment. In generalized estimating equation analysis, a gradual and significant increase of PCS was observed from patients not in remission (mean PCS 36.0) to remission on therapy (41.8) to remission off therapy (44.8). No significant difference in MCS was found between remission states. CONCLUSION we show a strong and persistent association between remission and PCS, but not MCS. These results support the relevance (construct validity) of the Definition of Remission in SLE remission definitions and the further development of a treat-to-target approach in SLE.
Collapse
Affiliation(s)
- Michel W P Tsang-A-Sjoe
- Department of Rheumatology, Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Irene E M Bultink
- Department of Rheumatology, Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Maaike Heslinga
- Department of Rheumatology, Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Lilian H van Tuyl
- Department of Rheumatology, Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Ronald F van Vollenhoven
- Department of Rheumatology, Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Alexandre E Voskuyl
- Department of Rheumatology, Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
61
|
Hosseini A, Gharibi T, Marofi F, Babaloo Z, Baradaran B. CTLA-4: From mechanism to autoimmune therapy. Int Immunopharmacol 2020; 80:106221. [PMID: 32007707 DOI: 10.1016/j.intimp.2020.106221] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 01/15/2020] [Accepted: 01/15/2020] [Indexed: 12/16/2022]
Abstract
CD28 and CTLA-4 are both important stimulatory receptors for the regulation of T cell activation. Because receptors share common ligands, B7.1 and B7.2, the expression and biological function of CTLA-4 is important for the negative regulation of T cell responses. Therefore, elimination of CTLA-4 can result in the breakdown of immune tolerance and the development of several diseases such as autoimmunity. Inhibitory signals of CTLA-4 suppress T cell responses and protect against autoimmune diseases in many ways. In this review, we summarize the structure, expression and signaling pathway of CTLA-4. We also highlight how CTLA-4 defends against potentially self-reactive T cells. Finally, we discuss how the CTLA-4 regulates a number of autoimmune diseases that indicate manipulation of this inhibitory molecule is a promise as a strategy for the immunotherapy of autoimmune diseases.
Collapse
Affiliation(s)
- Arezoo Hosseini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Babaloo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
62
|
Abstract
The role of inflammation in cardiovascular disease (CVD) is now widely accepted. Immune cells, including T cells, are influenced by inflammatory signals and contribute to the onset and progression of CVD. T cell activation is modulated by T cell co-stimulation and co-inhibition pathways. Immune checkpoint inhibitors (ICIs) targeting T cell inhibition pathways have revolutionized cancer treatment and improved survival in patients with cancer. However, ICIs might induce cardiovascular toxicity via T cell re-invigoration. With the rising use of ICIs for cancer treatment, a timely overview of the role of T cell co-stimulation and inhibition molecules in CVD is desirable. In this Review, the importance of these molecules in the pathogenesis of CVD is highlighted in preclinical studies on models of CVD such as vein graft disease, myocarditis, graft arterial disease, post-ischaemic neovascularization and atherosclerosis. This Review also discusses the therapeutic potential of targeting T cell co-stimulation and inhibition pathways to treat CVD, as well as the possible cardiovascular benefits and adverse events after treatment. Finally, the Review emphasizes that patients with cancer who are treated with ICIs should be monitored for CVD given the reported association between the use of ICIs and the risk of cardiovascular toxicity.
Collapse
|
63
|
Reynolds AC, Marshall NS, Hill CL, Adams RJ. Systematic review of the efficacy of commonly prescribed pharmacological treatments for primary treatment of sleep disturbance in patients with diagnosed autoimmune disease. Sleep Med Rev 2020; 49:101232. [PMID: 31911367 DOI: 10.1016/j.smrv.2019.101232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 11/04/2019] [Accepted: 11/04/2019] [Indexed: 01/19/2023]
Abstract
Sleep disturbances are commonly reported by patients with autoimmune disease, and are negatively related to both disease activity and quality of life. Despite the potential for sleep disturbance to exacerbate inflammatory pathways, acute management of sleep disturbance with pharmacological aids is not well understood in this patient group. The objective of this review was to determine the efficacy of pharmacological treatments for sleep disturbance to improve sleep outcomes in adult patients with diagnosed autoimmune disease. Four databases and grey literature were searched for randomized controlled trials which used a pharmacological treatment specifically to treat sleep disturbance in patients with diagnosed autoimmune disease, both in hospitalized and non-hospitalized settings. A sleep outcome was required to be the primary endpoint of the study. Of the 409 studies identified, a total of six were included in the systematic review. Risk of bias across the studies was largely unclear, making an assessment challenging; meta-analysis was not undertaken due to clinical and methodological heterogeneity between studies. While there appeared to be perceived improvement in self-reported sleep quantity and quality in existing studies with pharmacological treatment, there was also evidence of placebo effect on some measures. Relatively small numbers of patients have undergone gold-standard polysomnographic (PSG) recording of sleep which limits our knowledge of objectively determined sleep quantity and quality in patients with autoimmune disease receiving pharmacological treatment for sleep disturbance. Presently there is insufficient evidence to determine whether pharmacological treatment of sleep disturbance is beneficial for improving sleep quantity and quality in this patient group beyond rheumatoid arthritis.
Collapse
Affiliation(s)
- Amy C Reynolds
- The Appleton Institute, CQ University Australia, 44 Greenhill Road, Wayville, SA, Australia; School of Health, Medical and Applied Sciences, CQUniversity Australia Adelaide Campus, Wayville, SA, Australia.
| | - Nathaniel S Marshall
- CIRUS, Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, Susan Wakil School of Nursing and Midwifery, The University of Sydney, Sydney, NSW, Australia
| | - Catherine L Hill
- Rheumatology Unit, The Queen Elizabeth Hospital, Woodville, SA, Australia; Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Robert J Adams
- Adelaide Institute for Sleep Health: A Flinders Centre of Research Excellence, Flinders University, Bedford Park, SA, Australia; Respiratory and Sleep Service, Southern Adelaide Local Health Network, Bedford Park, SA, Australia
| |
Collapse
|
64
|
Khanna D, Spino C, Johnson S, Chung L, Whitfield M, Denton CP, Berrocal V, Jennifer F, Mehta B, Molitor J, Steen VD, Lafyatis R, Simms RW, Gill A, Kafaja S, Frech TM, Hsu V, Domsic RT, Pope JE, Gordon JK, Mayes MD, Schiopu E, Young A, Sandorfi N, Park J, Hant FN, Bernstein EJ, Chatterjee S, Castelino FV, Ajam A, Wang Y, Wood T, Allanore Y, Matucci-Cerinic M, Distler O, Singer O, Bush E, Fox D, Furst DE. Abatacept in Early Diffuse Cutaneous Systemic Sclerosis: Results of a Phase II Investigator-Initiated, Multicenter, Double-Blind, Randomized, Placebo-Controlled Trial. Arthritis Rheumatol 2020; 72:125-136. [PMID: 31342624 PMCID: PMC6935399 DOI: 10.1002/art.41055] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE T cells play a key role in the pathogenesis of early systemic sclerosis. This study was undertaken to assess the safety and efficacy of abatacept in patients with diffuse cutaneous systemic sclerosis (dcSSc). METHODS In this 12-month, randomized, double-blind, placebo-controlled trial, participants were randomized 1:1 to receive either subcutaneous abatacept 125 mg or matching placebo, stratified by duration of dcSSc. Escape therapy was allowed at 6 months for worsening disease. The coprimary end points were change in the modified Rodnan skin thickness score (MRSS) compared to baseline and safety over 12 months. Differences in longitudinal outcomes were assessed according to treatment using linear mixed models, with outcomes censored after initiation of escape therapy. Skin tissue obtained from participants at baseline was classified into intrinsic gene expression subsets. RESULTS Among 88 participants, the adjusted mean change in the MRSS at 12 months was -6.24 units for those receiving abatacept and -4.49 units for those receiving placebo, with an adjusted mean treatment difference of -1.75 units (P = 0.28). Outcomes for 2 secondary measures (Health Assessment Questionnaire disability index and a composite measure) were clinically and statistically significantly better with abatacept. The proportion of subjects in whom escape therapy was needed was higher in the placebo group relative to the abatacept group (36% versus 16%). In the inflammatory and normal-like skin gene expression subsets, decline in the MRSS over 12 months was clinically and significantly greater in the abatacept group versus the placebo group (P < 0.001 and P = 0.03, respectively). In the abatacept group, adverse events occurred in 35 participants versus 40 participants in the placebo group, including 2 deaths and 1 death, respectively. CONCLUSION In this phase II trial, abatacept was well-tolerated, but change in the MRSS was not statistically significant. Secondary outcome measures, including gene expression subsets, showed evidence in support of abatacept. These data should be confirmed in a phase III trial.
Collapse
Affiliation(s)
- Dinesh Khanna
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Scleroderma Program, University of Michigan, Ann Arbor, MI
| | - Cathie Spino
- Biostatistics, University of Michigan, Ann Arbor, MI
| | - Sindhu Johnson
- Rheumatology, Mount Sinai Hospital and University Health Network, Toronto, ON, Canada
| | - Lorinda Chung
- Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, CA
| | - Michael Whitfield
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
| | | | | | - Franks Jennifer
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
| | - Bhaven Mehta
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
| | - Jerry Molitor
- Rheumatic & Autoimmune Diseases, University of Minnesota, Minneapolis, MN
| | - Virginia D. Steen
- Rheumatology, MedStar Georgetown University Hospital, Washington, DC
| | - Robert Lafyatis
- Medicine/Division of Rheumatology, Pittsburgh University Medical Center, Pittsburgh, PA
| | - Robert W. Simms
- Rheumatology, Boston University School of Medicine, Boston, MA
| | - Anna Gill
- UCL Division of Medicine, Royal Free Campus, London, United Kingdom
| | - Suzanne Kafaja
- Department of Internal Medicine, University of California Los Angeles, David Geffen School of Medicine, Division of Rheumatology, Los Angeles, CA
| | - Tracy M. Frech
- Division of Rheumatology, University of Utah, Salt Lake City, UT
| | - Vivien Hsu
- Rheumatology, Robert Wood Johnson University Scleroderma Program, New Brunswick, NJ
| | - Robyn T. Domsic
- Medicine - Rheumatology, University of Pittsburgh, Pittsburgh, PA
| | - Janet E. Pope
- Department of Medicine, University of Western Ontario, London, ON, Canada
| | | | - Maureen D. Mayes
- Rheumatology, University of Texas McGovern Medical School, Houston, TX
| | - Elena Schiopu
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Scleroderma Program, University of Michigan, Ann Arbor, MI
| | - Amber Young
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Scleroderma Program, University of Michigan, Ann Arbor, MI
| | - Nora Sandorfi
- Perelman School of Medicine, University of Pennsylvania, Pittsburgh, PA
| | - Jane Park
- Seattle Rheumatology Associates, Seattle, WA
| | - Faye N. Hant
- Medicine/Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC
| | | | | | | | - Ali Ajam
- Division of Rheumatology-Immunology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Yue Wang
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
| | - Tammara Wood
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
| | | | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Ora Singer
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Scleroderma Program, University of Michigan, Ann Arbor, MI
| | - Erica Bush
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Scleroderma Program, University of Michigan, Ann Arbor, MI
| | - David Fox
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Scleroderma Program, University of Michigan, Ann Arbor, MI
| | | |
Collapse
|
65
|
Drug repurposing to improve treatment of rheumatic autoimmune inflammatory diseases. Nat Rev Rheumatol 2019; 16:32-52. [PMID: 31831878 DOI: 10.1038/s41584-019-0337-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2019] [Indexed: 02/08/2023]
Abstract
The past century has been characterized by intensive efforts, within both academia and the pharmaceutical industry, to introduce new treatments to individuals with rheumatic autoimmune inflammatory diseases (RAIDs), often by 'borrowing' treatments already employed in one RAID or previously used in an entirely different disease, a concept known as drug repurposing. However, despite sharing some clinical manifestations and immune dysregulation, disease pathogenesis and phenotype vary greatly among RAIDs, and limited understanding of their aetiology has made repurposing drugs for RAIDs challenging. Nevertheless, the past century has been characterized by different 'waves' of repurposing. Early drug repurposing occurred in academia and was based on serendipitous observations or perceived disease similarity, often driven by the availability and popularity of drug classes. Since the 1990s, most biologic therapies have been developed for one or several RAIDs and then tested among the others, with varying levels of success. The past two decades have seen data-driven repurposing characterized by signature-based approaches that rely on molecular biology and genomics. Additionally, many data-driven strategies employ computational modelling and machine learning to integrate multiple sources of data. Together, these repurposing periods have led to advances in the treatment for many RAIDs.
Collapse
|
66
|
Immune checkpoint molecules. Possible future therapeutic implications in autoimmune diseases. J Autoimmun 2019; 104:102333. [DOI: 10.1016/j.jaut.2019.102333] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
|
67
|
Magro R. Biological therapies and their clinical impact in the treatment of systemic lupus erythematosus. Ther Adv Musculoskelet Dis 2019; 11:1759720X19874309. [PMID: 31565077 PMCID: PMC6755633 DOI: 10.1177/1759720x19874309] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/12/2019] [Indexed: 12/15/2022] Open
Abstract
The development of biological therapies has had an impact on the management of several medical conditions. Their use in systemic lupus erythematosus (SLE), however, remains very limited. This review has summarized the evidence on the clinical effect of biologicals in SLE. Biological drugs with a number of targets have been studied in several phase II and III randomized controlled trials (RCTs). Positive results have been obtained in phase III RCTs with belimumab and this led to its license for active SLE. The clinical experience with belimumab has confirmed the efficacy and safety of belimumab in SLE. Promising results have been noted in phase II trials for blisibimod, sifalimumab, anifrolumab, and ustekinumab. Despite the fact that the RCTs with rituximab did not achieve their primary endpoint, clinical experience with rituximab is extensive and shows favorable clinical response in refractory renal and non-renal SLE. It is hoped that further ongoing phase III RCTs on a number of biological agents in SLE will highlight the potential role of other biologicals in the management of this challenging and heterogeneous condition.
Collapse
Affiliation(s)
- Rosalie Magro
- Rheumatology Department, Mater Dei Hospital, Tal-Qroqq, Msida, MSD 2090, Malta
| |
Collapse
|
68
|
Biologics in the Treatment of Lupus Erythematosus: A Critical Literature Review. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8142368. [PMID: 31396534 PMCID: PMC6668536 DOI: 10.1155/2019/8142368] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 06/18/2019] [Indexed: 01/07/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune inflammatory disease affecting multiple organ systems that runs an unpredictable course and may present with a wide variety of clinical manifestations. Advances in treatment over the last decades, such as use of corticosteroids and conventional immunosuppressive drugs, have improved life expectancy of SLE sufferers. Unfortunately, in many cases effective management of SLE is still related to severe drug-induced toxicity and contributes to organ function deterioration and infective complications, particularly among patients with refractory disease and/or lupus nephritis. Consequently, there is an unmet need for drugs with a better efficacy and safety profile. A range of different biologic agents have been proposed and subjected to clinical trials, particularly dedicated to this subset of patients whose disease is inadequately controlled by conventional treatment regimes. Unfortunately, most of these trials have given unsatisfactory results, with belimumab being the only targeted therapy approved for the treatment of SLE so far. Despite these pitfalls, several novel biologic agents targeting B cells, T cells, or cytokines are constantly being evaluated in clinical trials. It seems that they may enhance the therapeutic efficacy when combined with standard therapies. These efforts raise the hope that novel drugs for patients with refractory SLE may be available in the near future. This article reviews the current biological therapies being tested in the treatment of SLE.
Collapse
|
69
|
Czaja AJ. Immune inhibitory proteins and their pathogenic and therapeutic implications in autoimmunity and autoimmune hepatitis. Autoimmunity 2019; 52:144-160. [PMID: 31298041 DOI: 10.1080/08916934.2019.1641200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Key inhibitory proteins can blunt immune responses to self-antigens, and deficiencies in this repertoire may promote autoimmunity. The goals of this review are to describe the key immune inhibitory proteins, indicate their possible impact on the development of autoimmune disease, especially autoimmune hepatitis, and encourage studies to clarify their pathogenic role and candidacy as therapeutic targets. English abstracts were identified in PubMed by multiple search terms. Full length articles were selected for review, and secondary and tertiary bibliographies were developed. Cytotoxic T lymphocyte antigen-4 impairs ligation of CD28 to B7 ligands on antigen presenting cells and inhibits the adaptive immune response by increasing anti-inflammatory cytokines, generating regulatory T cells, and reducing T cell activation and proliferation. Programed cell death antigen-1 inhibits T cell selection, activation, and proliferation by binding with two ligands at different phases and locations of the immune response. A soluble alternatively spliced variant of this protein can dampen the inhibitory signal. Autoimmune hepatitis has been associated with polymorphisms of the cytotoxic T lymphocyte antigen-4 gene, reduced hepatic expression of a ligand of programed cell death antigen-1, an interfering soluble variant of this key inhibitory protein, and antibodies against it. Findings have been associated with laboratory indices of liver injury and suboptimal treatment response. Abatacept, belatacept, CD28 blockade, and induction of T cell exhaustion are management considerations that require scrutiny. In conclusion, deficiencies in key immune inhibitory proteins may promote the occurrence of autoimmune diseases, such as autoimmune hepatitis, and emerging interventions may overcome these deficiencies. Investigations should define the nature, impact and management of these inhibitory disturbances in autoimmune hepatitis.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| |
Collapse
|
70
|
Dall'Era M, Bruce IN, Gordon C, Manzi S, McCaffrey J, Lipsky PE. Current challenges in the development of new treatments for lupus. Ann Rheum Dis 2019; 78:729-735. [PMID: 30636212 DOI: 10.1136/annrheumdis-2018-214530] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/16/2018] [Accepted: 12/19/2018] [Indexed: 01/15/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a considerable impact on patients' quality of life. Despite the plethora of clinical trials for SLE since the turn of the millennium, only one new treatment has been approved for the condition, and the overall pace of successful drug development remains slow. Nevertheless, the myriad of clinical studies has yielded insights that have informed and refined our understanding of eligibility criteria, outcome measures and trial design in SLE. In this review, we highlight the achievements of clinical trials as well as the major pitfalls that have been identified in drug development for SLE and, in doing so, identify areas where collaboration and consensus will be important to facilitate progress.
Collapse
Affiliation(s)
- Maria Dall'Era
- Division of Rheumatology and Russell/Engleman Rheumatology Research Center, University of California San Francisco, San Francisco, California, USA
| | - Ian N Bruce
- Arthritis Research UK Centre for Epidemiology, Medicine and Health, The University of Manchester and NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Caroline Gordon
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, UK
| | - Susan Manzi
- Lupus Center of Excellence, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Janis McCaffrey
- Private Consultative Practice, West Vancouver, British Columbia, Canada
| | - Peter E Lipsky
- RILITE Research Institute, Charlottesville, Virginia, USA
| |
Collapse
|
71
|
Yasuda S. Emerging targets for the treatment of lupus erythematosus: There is no royal road to treating lupus. Mod Rheumatol 2019; 29:60-69. [PMID: 29947283 DOI: 10.1080/14397595.2018.1493909] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Systemic lupus erythematosus (SLE) is a highly heterogeneous autoimmune disease that preferentially affects women of child-bearing age. Most current treatments for SLE with the exception of belimumab are not target-specific. Nontargeted therapy such as corticosteroids, cyclophosphamide, and other immunosuppressive drugs results in unwanted adverse effects. Although progress in treatment, including supportive therapy, has dramatically improved the prognosis of patients with SLE, better treatment drugs and protocols with fewer adverse effects and higher efficacy for the most severe form of SLE are needed. Advancements in genomics, immunology, and pathophysiology in the field of systemic autoimmunity have provided physicians with increasing knowledge, but the most appropriate treatment for each patient with SLE remains to be established. Therefore, the search for novel treatment targets in patients with SLE is ongoing. This review focuses on recent findings in the genetics of lupus and the abnormalities in cellular interactions, cytokine profiles, and intracellular signaling in patients with SLE. Novel molecular targets for lupus, mostly introduced through clinical trials, are then discussed based on these findings.
Collapse
Affiliation(s)
- Shinsuke Yasuda
- a Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine , Hokkaido University , Sapporo , Japan
| |
Collapse
|
72
|
Barbacki A, Petri M, Aviña-Zubieta A, Alarcón GS, Bernatsky S. Fatigue Measurements in Systemic Lupus Erythematosus. J Rheumatol 2019; 46:1470-1477. [PMID: 30709953 DOI: 10.3899/jrheum.180831] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2019] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Fatigue is a frequent, disabling issue in systemic lupus erythematosus (SLE). It is, however, difficult to quantify. The Ad Hoc Committee on SLE Response Criteria for Fatigue in 2007 recommended using the Krupp Fatigue Severity Scale (FSS). Since then, the Functional Assessment of Chronic Illness Therapy (FACIT)-Fatigue Scale has also been validated in SLE. We performed a review of instruments used to measure fatigue in adult SLE patients from 2007 onward. METHODS We searched PubMed, Medline, and Embase (January 2008-October 2017), identifying clinical trials and observational studies in adult SLE, where fatigue was a specifically measured outcome. All English and French studies were reviewed to determine fatigue measures and results. RESULTS Thirty-seven studies met inclusion criteria. Eight scales were used. The visual analog scale (VAS), FSS, and FACIT-Fatigue Scale were most frequent. FSS was the most often used instrument in both clinical trials and observational studies. Twenty-five of the 37 studies demonstrated a difference in fatigue that was statistically significant and clinically meaningful. Of the 12 studies that did not, 6 used FSS, 3 used VAS, 2 used the Multidimensional Assessment of Fatigue, and 1 used the Brief Fatigue Index. All 6 studies using the FACIT-Fatigue Scale detected clinically meaningful and statistically significant differences. CONCLUSION VAS, FSS, and FACIT-Fatigue Scale were the most frequently used instruments in adult SLE studies from 2008 to 2017. Many studies detected clinically important changes in fatigue. Fatigue remains a key measure in both clinical trials and observational SLE studies.
Collapse
Affiliation(s)
- Ariane Barbacki
- From the Department of Medicine, Division of Rheumatology, McGill University Health Centre, Montreal, Quebec; Division of Rheumatology, Department of Medicine, University of British Columbia Faculty of Medicine, Vancouver, British Columbia; Arthritis Research Centre of Canada, Richmond, British Columbia; Department of Experimental Medicine, University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada; Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,A. Barbacki, MD, Department of Medicine, Division of Rheumatology, McGill University Health Centre; M. Petri, MD, MPH, Division of Rheumatology, Johns Hopkins University School of Medicine; A. Aviña-Zubieta, MD, MSc, PhD, Division of Rheumatology, Department of Medicine, and Department of Experimental Medicine, University of British Columbia Faculty of Medicine, and Arthritis Research Centre of Canada; G.S. Alarcón, MD, MPH, Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Alabama at Birmingham; S. Bernatsky, MD, PhD, Department of Medicine, Division of Rheumatology, McGill University Health Centre
| | - Michelle Petri
- From the Department of Medicine, Division of Rheumatology, McGill University Health Centre, Montreal, Quebec; Division of Rheumatology, Department of Medicine, University of British Columbia Faculty of Medicine, Vancouver, British Columbia; Arthritis Research Centre of Canada, Richmond, British Columbia; Department of Experimental Medicine, University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada; Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,A. Barbacki, MD, Department of Medicine, Division of Rheumatology, McGill University Health Centre; M. Petri, MD, MPH, Division of Rheumatology, Johns Hopkins University School of Medicine; A. Aviña-Zubieta, MD, MSc, PhD, Division of Rheumatology, Department of Medicine, and Department of Experimental Medicine, University of British Columbia Faculty of Medicine, and Arthritis Research Centre of Canada; G.S. Alarcón, MD, MPH, Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Alabama at Birmingham; S. Bernatsky, MD, PhD, Department of Medicine, Division of Rheumatology, McGill University Health Centre
| | - Antonio Aviña-Zubieta
- From the Department of Medicine, Division of Rheumatology, McGill University Health Centre, Montreal, Quebec; Division of Rheumatology, Department of Medicine, University of British Columbia Faculty of Medicine, Vancouver, British Columbia; Arthritis Research Centre of Canada, Richmond, British Columbia; Department of Experimental Medicine, University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada; Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,A. Barbacki, MD, Department of Medicine, Division of Rheumatology, McGill University Health Centre; M. Petri, MD, MPH, Division of Rheumatology, Johns Hopkins University School of Medicine; A. Aviña-Zubieta, MD, MSc, PhD, Division of Rheumatology, Department of Medicine, and Department of Experimental Medicine, University of British Columbia Faculty of Medicine, and Arthritis Research Centre of Canada; G.S. Alarcón, MD, MPH, Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Alabama at Birmingham; S. Bernatsky, MD, PhD, Department of Medicine, Division of Rheumatology, McGill University Health Centre
| | - Graciela S Alarcón
- From the Department of Medicine, Division of Rheumatology, McGill University Health Centre, Montreal, Quebec; Division of Rheumatology, Department of Medicine, University of British Columbia Faculty of Medicine, Vancouver, British Columbia; Arthritis Research Centre of Canada, Richmond, British Columbia; Department of Experimental Medicine, University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada; Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,A. Barbacki, MD, Department of Medicine, Division of Rheumatology, McGill University Health Centre; M. Petri, MD, MPH, Division of Rheumatology, Johns Hopkins University School of Medicine; A. Aviña-Zubieta, MD, MSc, PhD, Division of Rheumatology, Department of Medicine, and Department of Experimental Medicine, University of British Columbia Faculty of Medicine, and Arthritis Research Centre of Canada; G.S. Alarcón, MD, MPH, Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Alabama at Birmingham; S. Bernatsky, MD, PhD, Department of Medicine, Division of Rheumatology, McGill University Health Centre
| | - Sasha Bernatsky
- From the Department of Medicine, Division of Rheumatology, McGill University Health Centre, Montreal, Quebec; Division of Rheumatology, Department of Medicine, University of British Columbia Faculty of Medicine, Vancouver, British Columbia; Arthritis Research Centre of Canada, Richmond, British Columbia; Department of Experimental Medicine, University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada; Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA. .,A. Barbacki, MD, Department of Medicine, Division of Rheumatology, McGill University Health Centre; M. Petri, MD, MPH, Division of Rheumatology, Johns Hopkins University School of Medicine; A. Aviña-Zubieta, MD, MSc, PhD, Division of Rheumatology, Department of Medicine, and Department of Experimental Medicine, University of British Columbia Faculty of Medicine, and Arthritis Research Centre of Canada; G.S. Alarcón, MD, MPH, Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Alabama at Birmingham; S. Bernatsky, MD, PhD, Department of Medicine, Division of Rheumatology, McGill University Health Centre.
| |
Collapse
|
73
|
Leibler C, Thiolat A, Elsner RA, El Karoui K, Samson C, Grimbert P. Costimulatory blockade molecules and B-cell-mediated immune response: current knowledge and perspectives. Kidney Int 2019; 95:774-786. [PMID: 30711200 DOI: 10.1016/j.kint.2018.10.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/17/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022]
Abstract
There is an urgent need for therapeutic agents that target humoral alloimmunity in solid organ transplantation. This includes sensitized patients with preformed donor-specific human leukocyte antigen antibodies and patients who develop de novo donor-specific antibodies, both of which are associated with acute and chronic antibody-mediated rejection and allograft loss. In the last decade, both experimental and clinical studies highlighted the major impact of costimulation molecules in the control of immune responses both in the field of transplantation and autoimmune disease. Although these molecules have been initially developed to control the early steps of T-cell activation, recent evidence also supports their influence at several steps of the humoral response. In this review, we aim to provide an overview of the current knowledge of the effects of costimulatory blockade agents on humoral responses in both autoimmune and allogeneic contexts. We first present the effects of costimulatory molecules on the different steps of alloantibody production. We then summarize mechanisms and clinical results observed using cytotoxic T lymphocyte antigen-4 (CTLA4)-Ig molecules both in transplantation and autoimmunity. Finally, we present the potential interest and implications of other costimulatory family members as therapeutic targets, with emphasis on combinatorial approaches, for the optimal control of the alloantigen-specific humoral response.
Collapse
Affiliation(s)
- Claire Leibler
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Allan Thiolat
- Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Rebecca A Elsner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Khalil El Karoui
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Chloe Samson
- Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Philippe Grimbert
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France.
| |
Collapse
|
74
|
Guía de práctica clínica para el manejo del lupus eritematoso sistémico propuesta por el Colegio Mexicano de Reumatología. ACTA ACUST UNITED AC 2019; 15:3-20. [DOI: 10.1016/j.reuma.2018.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 12/31/2022]
|
75
|
Peschken CA, Wang Y, Abrahamowicz M, Pope J, Silverman E, Sayani A, Iczkovitz S, Ross J, Zummer M, Tucker L, Pineau C, Levy D, Hudson M, Hitchon CA, Huber AM, Smith CD, Avina-Zubieta A, Arbillaga H, Chédeville G, Wynant W, Fortin PR. Persistent Disease Activity Remains a Burden for Patients with Systemic Lupus Erythematosus. J Rheumatol 2018; 46:166-175. [DOI: 10.3899/jrheum.171454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2018] [Indexed: 12/26/2022]
Abstract
Objective.Persistent systemic lupus erythematosus (SLE) disease activity is associated with increased morbidity and mortality. In a multicenter cohort of patients with prevalent SLE, we described persistence, patterns, and predictors of change in disease activity over time.Methods.Based on SLE Disease Activity Index (SLEDAI)-2K scores at cohort entry, patients were classified into 4 groups: low (score < 4; LOW), moderate (4 to < 6; MOD), moderately high (6 to ≤ 10; MHIGH), and very high (> 10; VHIGH). Multivariable linear and longitudinal mixed linear regression models were used to identify predictors of change over time in SLEDAI-2K.Results.There were 2019 participants, with declining followup data over 5 years (1326, 580, 274, 186, and 148 patients, respectively). At cohort entry, mean (± SD) age was 42 (± 17) years, disease duration 11 (± 10) years, and 90% were female. The 4 groups included 44% LOW (n = 891), 20% MOD (n = 400), 22% MHIGH (n = 442), and 14% VHIGH (n = 286); therefore, 36% had clinically important SLE activity. The proportion of patients in the LOW group at entry who moved to a higher activity level varied from 30% (167/557) at 1 year, to 49% (41/83) at 3 years, and 54% (30/56) at 5 years. Among 181 patients with MOD to VHIGH entry activity and 3 years of followup, 116 (64.1%) remained active. In all analyses, only higher SLEDAI-2K at cohort entry remained a significant predictor of higher SLEDAI-2K in subsequent years.Conclusion.Higher SLEDAI-2K at study entry was the single major independent predictor of higher SLEDAI-2K over time, reflecting frequent persistence of active disease, even in patients with longstanding disease. This highlights gaps in the optimal treatment of SLE.
Collapse
|
76
|
Tayer-Shifman OE, Rosen CF, Wakani L, Touma Z. Novel biological therapeutic approaches to cutaneous lupus erythematosus. Expert Opin Biol Ther 2018; 18:1041-1047. [DOI: 10.1080/14712598.2018.1513484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
| | - Cheryl F. Rosen
- Division of Dermatology, Toronto Western Hospital, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Laura Wakani
- Centre For Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University of Toronto Lupus Clinic, Toronto, Canada
| | - Zahi Touma
- Centre For Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University of Toronto Lupus Clinic, Toronto, Canada
| |
Collapse
|
77
|
Izadi Z, Gandrup J, Katz PP, Yazdany J. Patient-reported outcome measures for use in clinical trials of SLE: a review. Lupus Sci Med 2018; 5:e000279. [PMID: 30167315 PMCID: PMC6109821 DOI: 10.1136/lupus-2018-000279] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/19/2018] [Accepted: 07/21/2018] [Indexed: 12/16/2022]
Abstract
Inclusion of patient-reported outcomes is important in SLE clinical trials as they allow capture of the benefits of a proposed intervention in areas deemed pertinent by patients. We aimed to compare the measurement properties of health-related quality of life (HRQoL) measures used in adults with SLE and to evaluate their responsiveness to interventions in randomised controlled trials (RCTs). A systematic review was undertaken using full original papers in English identified from three databases: MEDLINE, EMBASE and PubMed. Studies describing the validation of HRQoL measures in English-speaking adult patients with SLE and SLE drug RCTs that used an HRQoL measure were retrieved. Twenty-five validation papers and 26 RCTs were included in the indepth review evaluating the measurement properties of 4 generic (Medical Outcomes Study Short-Form 36 (SF36), Patient Reported Outcomes Measurement Information System (PROMIS) item-bank, EuroQol-5D, and Functional Assessment of Chronic Illness Therapy-Fatigue) and 3 disease-specific (Lupus Quality of Life (LupusQoL), Lupus Patient Reported Outcomes, Lupus Impact Tracker (LIT)) instruments. All measures had good convergent and discriminant validity. PROMIS provided the strongest evidence for known-group validity and reliability among generic instruments; however, data on its responsiveness have not been published. Across measures, standardised response means were generally indicative of poor-moderate sensitivity to longitudinal change. In RCTs, clinically important improvements were reported in SF36 scores from baseline; however, between-arm differences were frequently non-significant and non-important. SF36, PROMIS, LupusQoL and LIT had the strongest evidence for acceptable measurement properties, but few measures aside from the SF36 have been incorporated into clinical trials. This review highlights the importance of incorporating a broader range of SLE-specific HRQoL measures in RCTs and warrants further research that focuses on longitudinal responsiveness of newer instruments.
Collapse
Affiliation(s)
- Zara Izadi
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Julie Gandrup
- Division of Rheumatology, University of California, San Francisco, California, USA
| | - Patricia P Katz
- Division of Rheumatology, University of California, San Francisco, California, USA
| | - Jinoos Yazdany
- Division of Rheumatology, University of California, San Francisco, California, USA
| |
Collapse
|
78
|
Hu Y, Carman JA, Holloway D, Kansal S, Fan L, Goldstine C, Lee D, Somerville JE, Latek R, Townsend R, Johnsen A, Connolly S, Bandyopadhyay S, Shadick N, Weinblatt ME, Furie R, Nadler SG. Development of a Molecular Signature to Monitor Pharmacodynamic Responses Mediated by In Vivo Administration of Glucocorticoids. Arthritis Rheumatol 2018. [PMID: 29534336 PMCID: PMC6099349 DOI: 10.1002/art.40476] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To develop an objective, readily measurable pharmacodynamic biomarker of glucocorticoid (GC) activity. METHODS Genes modulated by prednisolone were identified from in vitro studies using peripheral blood mononuclear cells from normal healthy volunteers. Using the criteria of a >2-fold change relative to vehicle controls and an adjusted P value cutoff of less than 0.05, 64 up-regulated and 18 down-regulated genes were identified. A composite score of the up-regulated genes was generated using a single-sample gene set enrichment analysis algorithm. RESULTS GC gene signature expression was significantly elevated in peripheral blood leukocytes from normal healthy volunteers following oral administration of prednisolone. Expression of the signature increased in a dose-dependent manner, peaked at 4 hours postadministration, and returned to baseline levels by 48 hours after dosing. Lower expression was detected in normal healthy volunteers who received a partial GC receptor agonist, which is consistent with the reduced transactivation potential of this compound. In cohorts of patients with systemic lupus erythematosus and patients with rheumatoid arthritis, expression of the GC signature was negatively correlated with the percentages of peripheral blood lymphocytes and positively correlated with peripheral blood neutrophil counts, which is consistent with the known biology of the GC receptor. Expression of the signature largely agreed with reported GC use in these populations, although there was significant interpatient variability within the dose cohorts. CONCLUSION The GC gene signature identified in this study represents a pharmacodynamic marker of GC exposure.
Collapse
Affiliation(s)
- Yanhua Hu
- Bristol-Myers Squibb, Princeton, New Jersey
| | | | | | | | - Li Fan
- Bristol-Myers Squibb, Princeton, New Jersey
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Novelli R, Benigni A, Remuzzi G. The role of B7-1 in proteinuria of glomerular origin. Nat Rev Nephrol 2018; 14:589-596. [DOI: 10.1038/s41581-018-0037-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
80
|
Sciascia S, Radin M, Roccatello D, Sanna G, Bertolaccini ML. Recent advances in the management of systemic lupus erythematosus. F1000Res 2018; 7:F1000 Faculty Rev-970. [PMID: 30026918 PMCID: PMC6039948 DOI: 10.12688/f1000research.13941.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2018] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease presenting highly heterogeneous clinical manifestations and multi-systemic involvement. Patients are susceptible to relapse- and remission, thus making management challenging. Moreover, a considerable number of side effects may occur with conventional therapies; therefore, there is clearly a need for new therapeutic strategies. Since the pathogenesis of SLE is highly complex, it is far from being fully understood. However, greater understanding of the pathways and of the cellular and molecular mediators involved in SLE is being achieved. Emerging evidence has allowed the development of new biological therapeutic options targeting crucial molecular mediators involved in the pathogenesis of SLE. This literature review analyzes the availability of biological and target-directed treatments, phase II and III trials, and new therapies that are being developed for the treatment of SLE.
Collapse
Affiliation(s)
- Savino Sciascia
- Center of Research of Immunopathology and Rare Diseases, Coordinating Center of Piemonte and Valle d’Aosta Network for Rare Diseases, Department of Clinical and Biological Sciences, and SCDU Nephrology and Dialysis, S. Giovanni Bosco Hospital, Turin, Italy
| | - Massimo Radin
- Center of Research of Immunopathology and Rare Diseases, Coordinating Center of Piemonte and Valle d’Aosta Network for Rare Diseases, Department of Clinical and Biological Sciences, and SCDU Nephrology and Dialysis, S. Giovanni Bosco Hospital, Turin, Italy
| | - Dario Roccatello
- Center of Research of Immunopathology and Rare Diseases, Coordinating Center of Piemonte and Valle d’Aosta Network for Rare Diseases, Department of Clinical and Biological Sciences, and SCDU Nephrology and Dialysis, S. Giovanni Bosco Hospital, Turin, Italy
| | - Giovanni Sanna
- Louise Coote Lupus Unit, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Maria Laura Bertolaccini
- Academic Department of Vascular Surgery, School of Cardiovascular Medicine & Sciences, King’s College London, London, UK
| |
Collapse
|
81
|
Felten R, Dervovic E, Chasset F, Gottenberg JE, Sibilia J, Scher F, Arnaud L. The 2018 pipeline of targeted therapies under clinical development for Systemic Lupus Erythematosus: a systematic review of trials. Autoimmun Rev 2018; 17:781-790. [PMID: 29885544 DOI: 10.1016/j.autrev.2018.02.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 02/03/2018] [Indexed: 12/22/2022]
Abstract
Currently, Systemic Lupus Erythematosus (SLE) therapies range from antimalarials to glucocorticoids, in addition to immunosupressive agents or biologics such as rituximab or belimumab, when needed. Several unmet needs remain in the treatment SLE and more targeted drugs with improved safety profiles are expected. Based on recent advances in the understanding of the complex pathogenesis of SLE, several targeted treatments are currently assessed in clinical trials. In this study, we performed a systematic review of all targeted therapies under clinical development in SLE in 17 online registries of clinical trials. The search yielded a total of 1140 trials, from which we identified 74 targeted therapies for SLE. Those treatments target inflammatory cytokines, chemokines, or their receptors (n = 17), B cells or plasma cells (n = 17), intracellular signalling pathways (n = 10), T/B cells costimulation molecules (n = 8), interferons (n = 7), plasmacytoid dendritic cells (pDC) (n = 3), as well as various other targets (n = 12). Not all these candidate drugs will reach phase III, but the broad spectrum of drugs being investigated may satisfy the urgent need for improved lupus medications. The identification of biomarkers that would allow adequate prediction of response-to-therapy remains high, but when solved will allow a more rationale selection of the optimal pharmacological agent at the patient level.
Collapse
Affiliation(s)
- Renaud Felten
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, RESO, Laboratoire d'Immunopathologie et de Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR3572, France
| | - Elida Dervovic
- Service de Pharmacie-Stérilisation, Hôpitaux Universitaires de Strasbourg, France
| | - François Chasset
- Sorbonne Université, Faculté de Médecine Sorbonne Université, AP-HP, Service de Dermatologie et Allergologie, Hôpital Tenon, F-75020 Paris, France
| | - Jacques-Eric Gottenberg
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, RESO, Laboratoire d'Immunopathologie et de Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR3572, France
| | - Jean Sibilia
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, INSERM UMR_S1109, RESO, Université de Strasbourg, F-67000 Strasbourg, France
| | - Florence Scher
- Service de Pharmacie-Stérilisation, Hôpitaux Universitaires de Strasbourg, France
| | - Laurent Arnaud
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, INSERM UMR_S1109, RESO, Université de Strasbourg, F-67000 Strasbourg, France.
| |
Collapse
|
82
|
Ursini F, Russo E, De Giorgio R, De Sarro G, D'Angelo S. Current treatment options for psoriatic arthritis: spotlight on abatacept. Ther Clin Risk Manag 2018; 14:1053-1059. [PMID: 29922065 PMCID: PMC5995419 DOI: 10.2147/tcrm.s148586] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease of joints, tendon sheaths, and entheses affecting patients with established skin psoriasis, or, less frequently, patients without a personal history of psoriasis with a positive familial history. Many treatment options are now available to deal with the different aspects of the disease, including traditional and biological disease-modifying antirheumatic drugs and the recently released targeted synthetic disease-modifying antirheumatic drugs. However, ~40% of patients still fail to achieve a meaningful clinical response to first-line biologic therapy advocating the development of novel medications. It is now well accepted that T-cells participate in the immunopathogenesis of several autoimmune diseases. For this reason, the potential intervention on T-cells represented an attractive therapeutic target for a long time, becoming a clinical reality with the development of abatacept. Abatacept is a biologic agent selectively targeting the T-cell costimulatory signal delivered through the CD80/86-CD28 pathway and was approved in December 2005 by the US Food and Drug Administration and in May 2007 by European Medicines Agency for the treatment of patients with rheumatoid arthritis in combination with methotrexate. Based on the relevant role of T-cells in PsA pathogenesis and following the positive results obtained in a phase III clinical trial, abatacept recently received approval for treatment of patients with PsA. In this review, we will focus on the current knowledge about the emerging role of abatacept in treatment of PsA.
Collapse
Affiliation(s)
- Francesco Ursini
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy.,Associazione Calabrese per la Ricerca in Reumatologia, Catanzaro, Italy
| | - Emilio Russo
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | | | | | - Salvatore D'Angelo
- Rheumatology Department of Lucania, Rheumatology Institute of Lucania (IReL), Potenza Italy.,Basilicata Ricerca Biomedica (BRB), Potenza, Italy
| |
Collapse
|
83
|
Julià A, López-Longo FJ, Pérez Venegas JJ, Bonàs-Guarch S, Olivé À, Andreu JL, Aguirre-Zamorano MÁ, Vela P, Nolla JM, de la Fuente JLM, Zea A, Pego-Reigosa JM, Freire M, Díez E, Rodríguez-Almaraz E, Carreira P, Blanco R, Taboada VM, López-Lasanta M, Corbeto ML, Mercader JM, Torrents D, Absher D, Marsal S, Fernández-Nebro A. Genome-wide association study meta-analysis identifies five new loci for systemic lupus erythematosus. Arthritis Res Ther 2018; 20:100. [PMID: 29848360 PMCID: PMC5977506 DOI: 10.1186/s13075-018-1604-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/23/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a common systemic autoimmune disease with a complex genetic inheritance. Genome-wide association studies (GWAS) have significantly increased the number of significant loci associated with SLE risk. To date, however, established loci account for less than 30% of the disease heritability and additional risk variants have yet to be identified. Here we performed a GWAS followed by a meta-analysis to identify new genome-wide significant loci for SLE. METHODS We genotyped a cohort of 907 patients with SLE (cases) and 1524 healthy controls from Spain and performed imputation using the 1000 Genomes reference data. We tested for association using logistic regression with correction for the principal components of variation. Meta-analysis of the association results was subsequently performed on 7,110,321 variants using genetic data from a large cohort of 4036 patients with SLE and 6959 controls of Northern European ancestry. Genetic association was also tested at the pathway level after removing the effect of known risk loci using PASCAL software. RESULTS We identified five new loci associated with SLE at the genome-wide level of significance (p < 5 × 10- 8): GRB2, SMYD3, ST8SIA4, LAT2 and ARHGAP27. Pathway analysis revealed several biological processes significantly associated with SLE risk: B cell receptor signaling (p = 5.28 × 10- 6), CTLA4 co-stimulation during T cell activation (p = 3.06 × 10- 5), interleukin-4 signaling (p = 3.97 × 10- 5) and cell surface interactions at the vascular wall (p = 4.63 × 10- 5). CONCLUSIONS Our results identify five novel loci for SLE susceptibility, and biologic pathways associated via multiple low-effect-size loci.
Collapse
Affiliation(s)
- Antonio Julià
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain.
| | | | - José J Pérez Venegas
- Department of Rheumatology, Hospital del SAS de Jerez de la Frontera, 11407, Cádiz, Spain
| | - Silvia Bonàs-Guarch
- Barcelona Supercomputing Center. Joint BSC-CRG-IRB Research Program in Computational Biology, 08034, Barcelona, Spain
| | - Àlex Olivé
- Department of Rheumatology, Hospital Universitari Germans Trias i Pujol, 08916, Badalona, Spain
| | - José Luís Andreu
- Department of Rheumatology, Hospital Universitario Puerta de Hierro, 28222, Madrid, Spain
| | | | - Paloma Vela
- Department of Rheumatology, Hospital General Universitario de Alicante, 03010, Alicante, Spain
| | - Joan M Nolla
- Department of Rheumatology, Hospital Universitari de Bellvitge, 08907, Barcelona, Spain
| | | | - Antonio Zea
- Department of Rheumatology, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - José María Pego-Reigosa
- Department of Rheumatology, Hospital do Meixoeiro, Grupo IRIDIS, Instituto de Investigación sanitaria Galicia Sur (IISGS), 36312, Vigo, Spain
| | - Mercedes Freire
- Department of Rheumatology, Hospital Universitario A Coruña, 15006, A Coruña, Spain
| | - Elvira Díez
- Department of Rheumatology, Hospital Complejo Asistencial Universitario de León, 24071, León, Spain
| | | | - Patricia Carreira
- Department of Rheumatology, Hospital Universitario 12 de Octubre, 28041, Madrid, Spain
| | - Ricardo Blanco
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - Víctor Martínez Taboada
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - María López-Lasanta
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Mireia López Corbeto
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Josep M Mercader
- Barcelona Supercomputing Center. Joint BSC-CRG-IRB Research Program in Computational Biology, 08034, Barcelona, Spain
| | - David Torrents
- Barcelona Supercomputing Center. Joint BSC-CRG-IRB Research Program in Computational Biology, 08034, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Spain
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806, USA
| | - Sara Marsal
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain.
| | - Antonio Fernández-Nebro
- Department of Rheumatology, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga, 29011, Málaga, Spain
| |
Collapse
|
84
|
Pellerin L, Chen P, Gregori S, Hernandez-Hoyos G, Bacchetta R, Roncarolo MG. APVO210: A Bispecific Anti-CD86-IL-10 Fusion Protein (ADAPTIR™) to Induce Antigen-Specific T Regulatory Type 1 Cells. Front Immunol 2018; 9:881. [PMID: 29887861 PMCID: PMC5980965 DOI: 10.3389/fimmu.2018.00881] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/09/2018] [Indexed: 12/30/2022] Open
Abstract
IL-10 is a potent immunosuppressive cytokine that promotes the differentiation of tolerogenic dendritic cells (DC-10), and the subsequent induction of antigen-specific T regulatory type 1 (Tr1) cells, which suppress immune responses. However, IL-10 acts on multiple cell types and its effects are not solely inhibitory, therefore, limiting its use as immunomodulant. APVO210 is a bispecific fusion protein composed of an anti-CD86 antibody fused with monomeric IL-10 (ADAPTIR™ from Aptevo Therapeutics). APVO210 specifically induces IL-10R signaling in CD86+ antigen-presenting cells, but not in T and B cells. In this study, we tested whether APVO210 promotes the differentiation of tolerogenic DC-10 and the differentiation of antigen-specific CD4+ Tr1 cells in vitro. We compared the effect of APVO210 with that of recombinant human (rh) IL-10 on the in vitro differentiation of DC-10, induction of alloantigen-specific anergic CD4+ T cells, enrichment in CD49b+LAG3+ Tr1 cells mediating antigen-specific suppression, and stability upon exposure to inflammatory cytokines. APVO210 induced the differentiation of tolerogenic DC (DC-A210) that produced high levels of IL-10, expressed CD86, HLA-G, and intermediate levels of CD14 and CD16. These DC-A210 induced alloantigen-specific anergic T-cell cultures (T-alloA210) that were enriched in CD49b+ LAG3+ Tr1 cells, produced high levels of IL-10, and had suppressive properties. The phenotype and high IL-10 production by DC-A210, and the alloantigen-specific anergy of T-alloA210 were preserved upon exposure to the inflammatory cytokines IL-1β, IL-6, and TNF-α. The effects of APVO210 were comparable to that of dimeric rh IL-10. In conclusion, our data demonstrate that APVO210 drives the differentiation of tolerogenic DC and functional alloantigen-specific Tr1 cells in vitro. Since APVO210 specifically targets CD86+ cells, we hypothesize that it will specifically target CD86+ DC to induce Tr1 cells in vivo, and mediate antigen-specific immunological tolerance by induction of tolerogenic DC and Tr1 cells.
Collapse
Affiliation(s)
- Laurence Pellerin
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| | - Ping Chen
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Rosa Bacchetta
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| | - Maria Grazia Roncarolo
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
85
|
McCarthy EM, Sutton E, Nesbit S, White J, Parker B, Jayne D, Griffiths B, Isenberg DA, Rahman A, Gordon C, D'Cruz DP, Rhodes B, Lanyon P, Vital EM, Yee CS, Edwards CJ, Teh LS, Akil M, McHugh NJ, Zoma A, Bruce IN. Short-term efficacy and safety of rituximab therapy in refractory systemic lupus erythematosus: results from the British Isles Lupus Assessment Group Biologics Register. Rheumatology (Oxford) 2018; 57:470-479. [PMID: 29216396 PMCID: PMC5850287 DOI: 10.1093/rheumatology/kex395] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Indexed: 01/17/2023] Open
Abstract
Objectives To describe the baseline characteristics of SLE patients requiring biologic therapy in the UK and to explore short term efficacy and infection rates associated with rituximab (RTX) use. Methods Patients commencing biologic therapy for refractory SLE and who consented to join BILAG-BR were analysed. Baseline characteristics, disease activity (BILAG 2004/SLEDAI-2K) and rates of infection over follow-up were analysed. Response was defined as loss of all A and B BILAG scores to ⩽ 1 B score with no new A/B scores in other organ systems at 6 months. Results Two hundred and seventy SLE patients commenced biologic therapy from September 2010 to September 2015, most commonly RTX (n = 261). Two hundred and fifty (93%) patients were taking glucocorticoids at baseline at a median [interquartile range (IQR)] oral dose of 10 mg (5–20 mg) daily. Response rates at 6 months were available for 68% of patients. The median (IQR) BILAG score was 15 (10–23) at baseline and 3 (2–12) at 6 months (P < 0.0001). The median (IQR) SLEDAI-2K reduced from 8 (5–12) to 4 (0–7) (P < 0.001). Response was achieved in 49% of patients. There was also a reduction in glucocorticoid use to a median (IQR) dose of 7.5 mg (5–12 mg) at 6 months (P < 0.001). Serious infections occurred in 26 (10%) patients, being more frequent in the first 3 months post-RTX therapy. A higher proportion of early infections were non-respiratory (odds ratio = 1.98, 95% CI: 0.99, 3.9; P = 0.049). Conclusion RTX is safe and is associated with improvement in disease activity in refractory SLE patients with concomitant reductions in glucocorticoid use. Early vigilance for infection post-infusion is important to further improve treatment risks and benefits.
Collapse
Affiliation(s)
- Eoghan M McCarthy
- The Kellgren Centre for Rheumatology, NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester
| | - Emily Sutton
- Arthritis Research UK Centre for Epidemiology, University of Manchester, Manchester Academic Health Science Centre, Manchester
| | - Stephanie Nesbit
- Arthritis Research UK Centre for Epidemiology, University of Manchester, Manchester Academic Health Science Centre, Manchester
| | - James White
- Arthritis Research UK Centre for Epidemiology, University of Manchester, Manchester Academic Health Science Centre, Manchester
| | - Ben Parker
- The Kellgren Centre for Rheumatology, NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester.,Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester
| | - David Jayne
- Department of Medicine, Addenbooke's Hospital, Cambridge
| | | | - David A Isenberg
- Division of Rheumatology, University College London, Rayne Institute, London
| | - Anisur Rahman
- Division of Rheumatology, University College London, Rayne Institute, London
| | - Caroline Gordon
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham.,Rheumatology Department, City Hospital, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham
| | | | - Benjamin Rhodes
- Rheumatology Department, University Hospitals Birmingham NHS Foundation Trust, Old Queen Elizabeth Hospital, Birmingham
| | - Peter Lanyon
- Rheumatology Department, Nottingham University Hospitals NHS Trust, Nottingham
| | - Edward M Vital
- Leeds Institute for Rheumatic and Musculoskeletal Medicine, University of Leeds.,NIHR Leeds Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds
| | - Chee-Seng Yee
- Department of Rheumatology, Doncaster and Bassetlaw Hospitals NHS Foundation Trust, Doncaster
| | - Christopher J Edwards
- Musculoskeletal Research Unit, NIHR Wellcome Trust Clinical Research facility, The University of Southampton.,Department of Rheumatology, University Hospital Southampton NHS Foundation Trust, Southampton
| | - Lee-Suan Teh
- Department of Rheumatology, Royal Blackburn Hospital, Blackburn
| | - Mohammed Akil
- Rheumatology Department, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust
| | - Neil J McHugh
- Department of Rheumatology, Royal National Hospital for Rheumatic Diseases and Royal United Hospitals Bath NHS Foundation Trust, Bath, UK.,Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Asad Zoma
- Rheumatology Department, Hairmyres Hospital, Lanarkshire, UK
| | - Ian N Bruce
- The Kellgren Centre for Rheumatology, NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester University Hospitals NHS Foundation Trust, Manchester.,Arthritis Research UK Centre for Epidemiology, University of Manchester, Manchester Academic Health Science Centre, Manchester
| | | |
Collapse
|
86
|
|
87
|
Hofmann K, Clauder AK, Manz RA. Targeting B Cells and Plasma Cells in Autoimmune Diseases. Front Immunol 2018; 9:835. [PMID: 29740441 PMCID: PMC5924791 DOI: 10.3389/fimmu.2018.00835] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 04/05/2018] [Indexed: 12/29/2022] Open
Abstract
Success with B cell depletion using rituximab has proven the concept that B lineage cells represent a valid target for the treatment of autoimmune diseases, and has promoted the development of other B cell targeting agents. Present data confirm that B cell depletion is beneficial in various autoimmune disorders and also show that it can worsen the disease course in some patients. These findings suggest that B lineage cells not only produce pathogenic autoantibodies, but also significantly contribute to the regulation of inflammation. In this review, we will discuss the multiple pro- and anti-inflammatory roles of B lineage cells play in autoimmune diseases, in the context of recent findings using B lineage targeting therapies.
Collapse
Affiliation(s)
- Katharina Hofmann
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Schleswig-Holstein, Germany
| | - Ann-Katrin Clauder
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Schleswig-Holstein, Germany
| | - Rudolf Armin Manz
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Schleswig-Holstein, Germany
| |
Collapse
|
88
|
Merrill JT, Manzi S, Aranow C, Askanase A, Bruce I, Chakravarty E, Chong B, Costenbader K, Dall'Era M, Ginzler E, Hanrahan L, Kalunian K, Merola J, Raymond S, Rovin B, Saxena A, Werth VP. Lupus community panel proposals for optimising clinical trials: 2018. Lupus Sci Med 2018; 5:e000258. [PMID: 29657738 PMCID: PMC5894527 DOI: 10.1136/lupus-2018-000258] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/06/2018] [Accepted: 02/12/2018] [Indexed: 12/14/2022]
Abstract
Formidable impediments stand in the way of treatment development for lupus. These include the unwieldy size of current trials, international competition for scarce patients, complex outcome measures and a poor understanding of these outcomes in the world at large. The heterogeneity of the disease itself coupled to superimposition of variegated background polypharmacy has created enough immunological noise to virtually ensure the failure of lupus treatment trials, leaving an understandable suspicion that at least some of the results in testing failed drugs over the years may not have been negative, but merely uninterpretable. The authors have consulted with many clinical trial investigators, biopharmaceutical developers and stakeholders from government and voluntary sectors. This paper examines the available evidence that supports workable trial designs and proposes approaches to improve the odds of completing interpretable treatment development programs for lupus.
Collapse
Affiliation(s)
- Joan T Merrill
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Susan Manzi
- Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Cynthia Aranow
- Autoimmune and Musculoskeletal Disease, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Anca Askanase
- Columbia University Medical Center, New York City, New York, USA
| | - Ian Bruce
- University of Manchester, Manchester, England, UK
| | - Eliza Chakravarty
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Ben Chong
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Karen Costenbader
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Ellen Ginzler
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | - Leslie Hanrahan
- Research and Education, Lupus Foundation of America, Washington, District of Columbia, USA
| | - Ken Kalunian
- Rheumatology, University of California, San Diego, San Diego, USA
| | - Joseph Merola
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sandra Raymond
- Lupus Foundation of America, Washington, District of Columbia, USA
| | - Brad Rovin
- Internal Medicine/Nephrology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Amit Saxena
- Rheumatology, New York University, New York City, New York, USA
| | - Victoria P Werth
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
89
|
Abstract
Systemic lupus erythematosus (SLE) is a systemic disease of unknown aetiology with variable course and prognosis. Lupus nephritis (LN) is one of the important disease manifestations of SLE with considerable influence on patient outcomes. Immunosuppression therapy has made it possible to control the disease with improved life expectancy and quality of life. In the last few decades, various studies across the globe have clarified the role, dose and duration of immunosuppression currently in use and also provided evidence for new agents such as mycophenolate mofetil, calcineurin inhibitors and rituximab. However, there is still a need to develop new and specific therapy with less adverse effects. In this review, the current evidence of the treatment of LN and its evolution, and new classification criteria for SLE have been discussed. Also, rationale for low-dose intravenous cyclophosphamide as induction agent followed by azathioprine as maintenance agent has been provided with emphasis on individualized and holistic approach.
Collapse
Affiliation(s)
- Ajay Jaryal
- Department of Nephrology, Indira Gandhi Medical College (IGMC), Shimla, India
| | - Sanjay Vikrant
- Department of Nephrology, Indira Gandhi Medical College (IGMC), Shimla, India
| |
Collapse
|
90
|
Pollmann R, Schmidt T, Eming R, Hertl M. Pemphigus: a Comprehensive Review on Pathogenesis, Clinical Presentation and Novel Therapeutic Approaches. Clin Rev Allergy Immunol 2018; 54:1-25. [DOI: 10.1007/s12016-017-8662-z] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
91
|
Danion F, Sparsa L, Arnaud L, Alsaleh G, Lefebvre F, Gies V, Martin T, Lukas C, Durckel J, Ardizzone M, Javier RM, Kleinmann JF, Moreau P, Blaison G, Goetz J, Chatelus E, Gottenberg JE, Sibilia J, Sordet C. Long-term efficacy and safety of antitumour necrosis factor alpha treatment in rhupus: an open-label study of 15 patients. RMD Open 2017; 3:e000555. [PMID: 29435362 PMCID: PMC5761296 DOI: 10.1136/rmdopen-2017-000555] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/04/2017] [Accepted: 10/21/2017] [Indexed: 11/03/2022] Open
Abstract
Background The efficacy of antitumour necrosis factor alpha (anti-TNF-α) treatment is well recognised in rheumatoid arthritis (RA) but remains controversial in systemic lupus erythematosus (SLE). Therefore, the role of anti-TNF-α treatment in 'Rhupus', a disease sharing features of RA and SLE, is still debated. Objective To evaluate the efficacy and tolerance of anti-TNF-α in patients with rhupus. Methods Fifteen patients with rhupus with Disease Activity Score 28 (DAS 28) >3.2 despite conventional disease-modifying anti-rheumatic drugs were included in an open-label study. Patients were monitored at months (M) 3, 6, 12, 24 and 60 with SLE Disease Activity Index (SLEDAI) and DAS 28. Statistical analyses were performed using Bayesian methods and Prob >97.5% was considered significant. Results Twelve patients were treated with etanercept for a median duration of 62.5 (range: 6-112) months and three patients by adalimumab during 36.0 (range: 4-52) months. At baseline, median DAS 28 and SLEDAI were 5.94 (4.83-8.09) and 6 (4-8), respectively. DAS 28 and SLEDAI decreased significantly after 3 months, respectively, to 3.70 (1.80-6.42) and 4 (0-6) (Prob >99.9%, for both). These changes persisted at M6, M12, M24 and M60 (Prob >99.9%, for all). Median prednisone dose decreased significantly from 15 (5-35) mg/day to 5 (0-20) mg/day after 6 months and over the follow-up (Prob >99.9%, for all). Tolerance was acceptable, with a severe infection rate of 3.0 per 100 patient-years. Conclusion This pilot study suggests that anti-TNF-α is effective in patients with rhupus with refractive arthritis and has an acceptable safety profile.
Collapse
Affiliation(s)
- François Danion
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Laetitia Sparsa
- Department of Rheumatology, Centre de compétence maladies rares et autoimmunes, Centre Hospitalier de Mulhouse, Mulhouse, France
| | - Laurent Arnaud
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Ghada Alsaleh
- UMR 1109 Immuno-Rhumatologie Moléculaire, INSERM, Strasbourg, France
| | - François Lefebvre
- Department of Public Health, Hopitaux universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Vincent Gies
- UPR 3572 Immunopathology and Therapeutic Chemistry, CNRS, Strasbourg, France
| | - Thierry Martin
- UPR 3572 Immunopathology and Therapeutic Chemistry, CNRS, Strasbourg, France.,Department of Internal Medicine, Centre de références des maladies rares et auto immunes, Hopitaux universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Cédric Lukas
- Department of Rheumatology, Lapeyronie Hospital, Montpellier University, Montpellier, France
| | - Jean Durckel
- Department of Radiology, Hopitaux universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Marc Ardizzone
- Department of Rheumatology, Centre de compétence maladies rares et autoimmunes, Centre Hospitalier de Mulhouse, Mulhouse, France
| | - Rose-Marie Javier
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Jean-François Kleinmann
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Paul Moreau
- Department of Rheumatology, Hopital Louis Pasteur, Colmar, Alsace, France
| | - Gilles Blaison
- Department of Internal Medicine, Hopital Louis Pasteur, Colmar, Alsace, France
| | - Joelle Goetz
- Department of Immunology, Hopitaux universitaires de Strasbourg, Strasbourg, France
| | - Emmanuel Chatelus
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Jacques-Eric Gottenberg
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France.,UPR 3572 Immunopathology and Therapeutic Chemistry, CNRS, Strasbourg, France
| | - Jean Sibilia
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France.,UMR 1109 Immuno-Rhumatologie Moléculaire, INSERM, Strasbourg, France
| | - Christelle Sordet
- Department of Rheumatology, Centre de référence des maladies rares et auto immunes, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
92
|
Vanhove B, Poirier N, Fakhouri F, Laurent L, 't Hart B, Papotto PH, Rizzo LV, Zaitsu M, Issa F, Wood K, Soulillou JP, Blancho G. Antagonist Anti-CD28 Therapeutics for the Treatment of Autoimmune Disorders. Antibodies (Basel) 2017; 6:antib6040019. [PMID: 31548534 PMCID: PMC6698823 DOI: 10.3390/antib6040019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/16/2017] [Accepted: 11/18/2017] [Indexed: 12/17/2022] Open
Abstract
The effector functions of T lymphocytes are responsible for most autoimmune disorders and act by directly damaging tissues or by indirectly promoting inflammation and antibody responses. Co-stimulatory and co-inhibitory T cell receptor molecules are the primary pharmacological targets that enable interference with immune-mediated diseases. Among these, selective CD28 antagonists have drawn special interest, since they tip the co-stimulation/co-inhibition balance towards efficiently inhibiting effector T cells while promoting suppression by pre-existing regulatory T-cells. After having demonstrated outstanding therapeutic efficacy in multiple models of autoimmunity, inflammation and transplantation, and safety in phase-I studies in humans, selective CD28 antagonists are currently in early clinical development for the treatment of systemic lupus erythematous and rheumatoid arthritis. Here, we review the available proof of concept studies for CD28 antagonists in autoimmunity, with a special focus on the mechanisms of action.
Collapse
Affiliation(s)
- Bernard Vanhove
- OSE Immunotherapeutics, 44200 Nantes, France.
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| | - Nicolas Poirier
- OSE Immunotherapeutics, 44200 Nantes, France.
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| | - Fadi Fakhouri
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| | - Laetitia Laurent
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
| | - Bert 't Hart
- Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands.
- Department Neuroscience, University of Groningen, University Medical Center, 9713 GZ Groningen, The Netherlands.
| | - Pedro H Papotto
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal.
| | - Luiz V Rizzo
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627-701, 2-SS Bloco A, 05651-901 São Paulo, Brazil.
| | - Masaaki Zaitsu
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| | - Kathryn Wood
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| | - Jean-Paul Soulillou
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
| | - Gilles Blancho
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, 44035 Nantes, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France.
| |
Collapse
|
93
|
Borba HHL, Funke A, Wiens A, Utiyama SRDR, Perlin CM, Pontarolo R. Update on Biologic Therapies for Systemic Lupus Erythematosus. Curr Rheumatol Rep 2017; 18:44. [PMID: 27299782 DOI: 10.1007/s11926-016-0589-5] [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] [Indexed: 12/30/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic multisystemic autoimmune disease driven by genetic, hormonal, and environmental factors. Despite the advances in diagnostic and therapeutic approaches in the last decades, SLE still leads to significant morbidity and increased mortality. Although a cure for SLE is still unknown, treatment is required to control acute disease exacerbation episodes (flares), decrease the frequency and severity of subsequent lupus flares, address comorbidities, and prevent end-organ damage. While conventional SLE pharmacotherapy may exhibit suboptimal efficacy and substantial toxicity, a growing knowledge of the disease pathogenesis enabled the research on novel therapeutic agents directed at specific disease-related targets. In this paper, we review the recent progress in the clinical investigation of biologic agents targeting B cells, T cells, cytokines, innate immunity, and other immunologic or inflammatory pathways. Although many investigational agents exhibited insufficient efficacy or inadequate safety in clinical trials, one of them, belimumab, fulfilled the efficacy and safety regulatory requirements and was approved for the treatment of SLE in Europe and the USA, which confirms that, despite all difficulties, advances in this field are possible.
Collapse
Affiliation(s)
- Helena Hiemisch Lobo Borba
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Research Program, Federal University of Parana, Campus III, Av. Pref. Lothario Meissner, 632, Jardim Botanico, Curitiba, PR, 80210-170, Brazil
| | - Andreas Funke
- Rheumatology Service, Hospital de Clinicas, Federal University of Parana, Curitiba, PR, Brazil
| | - Astrid Wiens
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Research Program, Federal University of Parana, Campus III, Av. Pref. Lothario Meissner, 632, Jardim Botanico, Curitiba, PR, 80210-170, Brazil
| | - Shirley Ramos da Rosa Utiyama
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Research Program, Federal University of Parana, Campus III, Av. Pref. Lothario Meissner, 632, Jardim Botanico, Curitiba, PR, 80210-170, Brazil
| | - Cássio Marques Perlin
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Research Program, Federal University of Parana, Campus III, Av. Pref. Lothario Meissner, 632, Jardim Botanico, Curitiba, PR, 80210-170, Brazil
| | - Roberto Pontarolo
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Research Program, Federal University of Parana, Campus III, Av. Pref. Lothario Meissner, 632, Jardim Botanico, Curitiba, PR, 80210-170, Brazil.
| |
Collapse
|
94
|
Abstract
The immune system is guided by a series of checks and balances, a major component of which is a large array of co-stimulatory and co-inhibitory pathways that modulate the host response. Although co-stimulation is essential for boosting and shaping the initial response following signaling through the antigen receptor, inhibitory pathways are also critical for modulating the immune response. Excessive co-stimulation and/or insufficient co-inhibition can lead to a breakdown of self-tolerance and thus to autoimmunity. In this review, we will focus on the role of co-stimulatory and co-inhibitory pathways in two systemic (systemic lupus erythematosus and rheumatoid arthritis) and two organ-specific (multiple sclerosis and type 1 diabetes) emblematic autoimmune diseases. We will also discuss how mechanistic analysis of these pathways has led to the identification of potential therapeutic targets and initiation of clinical trials for autoimmune diseases, as well as outline some of the challenges that lie ahead.
Collapse
Affiliation(s)
- Qianxia Zhang
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA.
| |
Collapse
|
95
|
|
96
|
Giacomelli R, Afeltra A, Alunno A, Baldini C, Bartoloni-Bocci E, Berardicurti O, Carubbi F, Cauli A, Cervera R, Ciccia F, Cipriani P, Conti F, De Vita S, Di Benedetto P, Doria A, Drosos AA, Favalli EG, Gandolfo S, Gatto M, Grembiale RD, Liakouli V, Lories R, Lubrano E, Lunardi C, Margiotta DPE, Massaro L, Meroni P, Minniti A, Navarini L, Pendolino M, Perosa F, Pers JO, Prete M, Priori R, Puppo F, Quartuccio L, Ruffatti A, Ruscitti P, Russo B, Sarzi-Puttini P, Shoenfeld Y, Somarakis GA, Spinelli FR, Tinazzi E, Triolo G, Ursini F, Valentini G, Valesini G, Vettori S, Vitali C, Tzioufas AG. International consensus: What else can we do to improve diagnosis and therapeutic strategies in patients affected by autoimmune rheumatic diseases (rheumatoid arthritis, spondyloarthritides, systemic sclerosis, systemic lupus erythematosus, antiphospholipid syndrome and Sjogren's syndrome)? Autoimmun Rev 2017; 16:911-924. [DOI: 10.1016/j.autrev.2017.07.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 05/20/2017] [Indexed: 02/06/2023]
|
97
|
Kubo S, Nakayamada S, Yoshikawa M, Miyazaki Y, Sakata K, Nakano K, Hanami K, Iwata S, Miyagawa I, Saito K, Tanaka Y. Peripheral Immunophenotyping Identifies Three Subgroups Based on T Cell Heterogeneity in Lupus Patients. Arthritis Rheumatol 2017; 69:2029-2037. [DOI: 10.1002/art.40180] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 06/08/2017] [Indexed: 12/29/2022]
Affiliation(s)
- Satoshi Kubo
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - Shingo Nakayamada
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - Maiko Yoshikawa
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - Yusuke Miyazaki
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - Kei Sakata
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - Kazuhisa Nakano
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - Kentaro Hanami
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - Shigeru Iwata
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - Ippei Miyagawa
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - Kazuyoshi Saito
- University of Occupational and Environmental Health; Kitakyushu Japan
| | - Yoshiya Tanaka
- University of Occupational and Environmental Health; Kitakyushu Japan
| |
Collapse
|
98
|
Adams AB, Ford ML, Larsen CP. Costimulation Blockade in Autoimmunity and Transplantation: The CD28 Pathway. THE JOURNAL OF IMMUNOLOGY 2017; 197:2045-50. [PMID: 27591335 DOI: 10.4049/jimmunol.1601135] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 07/11/2016] [Indexed: 12/30/2022]
Abstract
T cell activation is a complex process that requires multiple cell signaling pathways, including a primary recognition signal and additional costimulatory signals. TCR signaling in the absence of costimulatory signals can lead to an abortive attempt at activation and subsequent anergy. One of the best-characterized costimulatory pathways includes the Ig superfamily members CD28 and CTLA-4 and their ligands CD80 and CD86. The development of the fusion protein CTLA-4-Ig as an experimental and subsequent therapeutic tool is one of the major success stories in modern immunology. Abatacept and belatacept are clinically approved agents for the treatment of rheumatoid arthritis and renal transplantation, respectively. Future interventions may include selective CD28 blockade to block the costimulatory potential of CD28 while exploiting the coinhibitory effects of CTLA-4.
Collapse
Affiliation(s)
- Andrew B Adams
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Mandy L Ford
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Christian P Larsen
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
99
|
Bandyopadhyay S, Connolly SE, Jabado O, Ye J, Kelly S, Maldonado MA, Westhovens R, Nash P, Merrill JT, Townsend RM. Identification of biomarkers of response to abatacept in patients with SLE using deconvolution of whole blood transcriptomic data from a phase IIb clinical trial. Lupus Sci Med 2017; 4:e000206. [PMID: 29214034 PMCID: PMC5704740 DOI: 10.1136/lupus-2017-000206] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 06/05/2017] [Accepted: 06/11/2017] [Indexed: 12/13/2022]
Abstract
Objective To characterise patients with active SLE based on pretreatment gene expression-defined peripheral immune cell patterns and identify clusters enriched for potential responders to abatacept treatment. Methods This post hoc analysis used baseline peripheral whole blood transcriptomic data from patients in a phase IIb trial of intravenous abatacept (~10 mg/kg/month). Cell-specific genes were used with a published deconvolution algorithm to identify immune cell proportions in patient samples, and unsupervised consensus clustering was generated. Efficacy data were re-analysed. Results Patient data (n=144: abatacept: n=98; placebo: n=46) were grouped into four main clusters (C) by predominant characteristic cells: C1—neutrophils; C2—cytotoxic T cells, B-cell receptor-ligated B cells, monocytes, IgG memory B cells, activated T helper cells; C3—plasma cells, activated dendritic cells, activated natural killer cells, neutrophils; C4—activated dendritic cells, cytotoxic T cells. C3 had the highest baseline total British Isles Lupus Assessment Group (BILAG) scores, highest antidouble-stranded DNA autoantibody levels and shortest time to flare (TTF), plus trends in favour of response to abatacept over placebo: adjusted mean difference in BILAG score over 1 year, −4.78 (95% CI −12.49 to 2.92); median TTF, 56 vs 6 days; greater normalisation of complement component 3 and 4 levels. Differential improvements with abatacept were not seen in other clusters, except for median TTF in C1 (201 vs 109 days). Conclusions Immune cell clustering segmented disease severity and responsiveness to abatacept. Definition of immune response cell types may inform design and interpretation of SLE trials and treatment decisions. Trial registration number NCT00119678; results.
Collapse
Affiliation(s)
| | - Sean E Connolly
- US Medical, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Omar Jabado
- Translational Bioinformatics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - June Ye
- Global Biometric Sciences, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Sheila Kelly
- US Medical, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | | | - Rene Westhovens
- Department of Development and Regeneration KU Leuven, Skeletal Biology and Engineering Research Center; Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Peter Nash
- Department of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Joan T Merrill
- Arthritis and Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Robert M Townsend
- Clinical Biomarkers, Bristol-Myers Squibb, Princeton, New Jersey, USA
| |
Collapse
|
100
|
Abstract
PURPOSE OF REVIEW With advancement in our understanding of pathogenic mechanisms in systemic lupus erythematosus (SLE), there is tremendous enthusiasm in examining drugs, old and new, to improve outcomes. This review highlights recent trials' successes and impasses that have come to fore. RECENT FINDINGS Among B-cell therapies, belimumab continues its run of successes with sustained safety and tolerability documented in a long-term extension as well as the likely approval of a subcutaneous formulation in the near future. With greater antibody-dependent cytotoxicity and less immunogenicity, there is hope for obinituzumab to succeed where its anti-CD 20 predecessors have failed. Drugs targeting type I interferons - sifalimumab and anifrolumab - have been efficacious albeit with an increase in incidence of Herpes zoster infections. There is also renewed interest in evaluating the efficacy of calcineurin inhibitors, specifically tacrolimus in the induction and maintenance of lupus nephritis. Introspection into clinical trial designs have highlighted the effects of entry criteria, end points, background medications and geographical differences on study outcomes. SUMMARY There are at least 50 drugs and targets being evaluated in SLE. In addition to developing new drugs to treat lupus, future trials have to focus on more effective study designs to improve chances of trial success.
Collapse
|