51
|
Hong JH, Kim HJ, Park KS, Ku BJ. Paradigm shift in the management of type B insulin resistance. ANNALS OF TRANSLATIONAL MEDICINE 2019; 6:S98. [PMID: 30740419 DOI: 10.21037/atm.2018.11.21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jun Hwa Hong
- Department of Internal Medicine, College of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Hyun Jin Kim
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Kang Seo Park
- Department of Internal Medicine, College of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Bon Jeong Ku
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
52
|
Bartsch T, Rempe T, Leypoldt F, Riedel C, Jansen O, Berg D, Deuschl G. The spectrum of progressive multifocal leukoencephalopathy: a practical approach. Eur J Neurol 2019; 26:566-e41. [PMID: 30629326 DOI: 10.1111/ene.13906] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 01/08/2019] [Indexed: 12/21/2022]
Abstract
John Cunningham virus (JCV) infection of the central nervous system causes progressive multifocal leukoencephalopathy (PML) in patients with systemic immunosuppression. With the increased application of modern immunotherapy and biologics in various immune-mediated disorders, the PML risk spectrum has changed. Thus, new tools and strategies for risk assessment and stratification in drug-associated PML such as the JCV antibody indices have been introduced. Imaging studies have highlighted atypical presentations of cerebral JCV disease such as granule cell neuronopathy. Imaging markers have been developed to differentiate PML from new multiple sclerosis lesions and to facilitate the early identification of pre-clinical manifestations of PML and its immune reconstitution inflammatory syndrome. PML can be diagnosed either by brain biopsy or by clinical, radiographic and virological criteria. Experimental treatment options including immunization and modulation of interleukin-mediated immune response are emerging. PML should be considered in any patient with compromised systemic or central nervous system immune surveillance presenting with progressive neurological symptoms.
Collapse
Affiliation(s)
- T Bartsch
- Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - T Rempe
- Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Department of Neurology, University of Florida, Gainesville, FL, USA
| | - F Leypoldt
- Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Department of Neuroimmunology, Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - C Riedel
- Institute of Neuroradiology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - O Jansen
- Institute of Neuroradiology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - D Berg
- Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - G Deuschl
- Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
53
|
Abstract
B cells play a vital function in multiple sclerosis (MS) pathogenesis through an array of effector functions. All currently approved MS disease-modifying therapies alter the frequency, phenotype, or homing of B cells in one way or another. The importance of this mechanism of action has been reinforced with the successful development and clinical testing of B-cell-depleting monoclonal antibodies that target the CD20 surface antigen. Ocrelizumab, a humanized anti-CD20 monoclonal antibody, was approved by the Food and Drug Administration (FDA) in March 2017 after pivotal trials showed dramatic reductions in inflammatory disease activity in relapsing MS as well as lessening of disability progression in primary progressive MS. These and other clinical studies place B cells at the center of the inflammatory cascade in MS and provide a launching point for development of therapies that target selective pathogenic B-cell populations.
Collapse
Affiliation(s)
- Joseph J Sabatino
- Multiple Sclerosis Center, Department of Neurology, University of California, San Francisco, California 94158
| | - Scott S Zamvil
- Multiple Sclerosis Center, Department of Neurology, University of California, San Francisco, California 94158
| | - Stephen L Hauser
- Multiple Sclerosis Center, Department of Neurology, University of California, San Francisco, California 94158
| |
Collapse
|
54
|
Shaygannejad V, Fayyazi E, Badihian S, Mirmosayyeb O, Manouchehri N, Ashtari F, Asgari N. Long-term tolerability, safety and efficacy of rituximab in neuromyelitis optica spectrum disorder: a prospective study. J Neurol 2019; 266:642-650. [PMID: 30635724 DOI: 10.1007/s00415-019-09180-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/29/2018] [Accepted: 01/02/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Neuromyelitis optica spectrum disorder (NMOSD) is a B-cell-mediated disease with autoimmunity towards the astrocyte water channel aquaporin-4 (AQP-4) in the central nervous system. OBJECTIVE To assess the long-term safety and efficacy in NMOSD patients receiving maintenance therapy with B-cell-depleting agent rituximab for more than 2 years. METHOD NMOSD patients were included prospectively from 2014 to 2018 and received continuous cycles of rituximab infusions biannually. Incidence of adverse events (AE), serious AEs (SAE), and infusion-related AEs were evaluated through monthly phone calls and neurological examination every 4 months. RESULTS A total of 44 NMOSD patients were included, of those 30 were treatment naive (68%). The mean age was 37.2 years with 79.5% females. With overall observation period of 31.6 ± 7.3 months (24-48 months), tolerability was assessed as satisfactory in most cases. We observed infusion reactions (mostly mild) in 31.8% of patients and 31.8% never experienced any AEs after a mean 5.1 cycles of rituximab therapy. Rituximab was also beneficial in terms of improvement in relapse rate (from 0.26 ± 0.54 to 0, P = 0.003) and Expanded Disability Status Scale (from 4.1 ± 1.8 to 3.1 ± 1.8, P < 0.001). Stratification according to AQP4-IgG serostatus showed no difference between groups. CONCLUSION Rituximab treatment is well tolerated, safe, and efficacious with a minor risk of mild infusion reactions for NMOSD patients.
Collapse
Affiliation(s)
- V Shaygannejad
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - E Fayyazi
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - S Badihian
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - O Mirmosayyeb
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - N Manouchehri
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - F Ashtari
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - N Asgari
- Department of Neurobiology, Institute of Molecular Medicine, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
55
|
Safety and Efficacy of Rituximab in Multiple Sclerosis: A Retrospective Observational Study. J Immunol Res 2018; 2018:9084759. [PMID: 30539030 PMCID: PMC6260423 DOI: 10.1155/2018/9084759] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/31/2018] [Accepted: 09/16/2018] [Indexed: 12/18/2022] Open
Abstract
Objective To evaluate the efficacy and safety of rituximab in multiple sclerosis in a clinical practice setting. Methods Clinical data for all adult patients with multiple sclerosis (MS) treated with off-label rituximab at a single MS center in Lebanon between March 2008 and April 2017 were retrospectively collected from medical charts. The main efficacy outcomes assessed were annualized relapse rate (ARR) and proportion of patients free from relapses, disability progression, or magnetic resonance imaging (MRI) activity. Results A total of 89 rituximab-treated patients were included: 59 relapsing-remitting MS (RRMS) and 30 progressive MS (PMS). Patients were treated with 1000 or 2000 mg rituximab IV every 6-12 months for a mean duration of 22.2 ± 24.8 months. The subjects were 65.2% females with a mean age of 40.5 ± 12.3 years and a mean disease duration of 7.9 ± 6.2 years. During treatment, the ARR decreased from 1.07 at baseline to 0.11 in RRMS (p < 0.0001) and from 0.25 to 0.16 in PMS patients (p = 0.593). The mean Expanded Disability Status Scale (EDSS) remained unchanged in both RRMS and PMS patients. Between baseline and the last follow-up, the percent of patients free from any new MRI lesions increased from 18.6% to 92.6% in the RRMS group and from 43.3% to 82% in the PMS group. No evidence of disease activity (NEDA) was achieved in 74% of patients at 1 year of treatment. A total of 64 adverse events (AEs) (71.9%) were recorded with the most common being infusion-related reactions in 25.8% of patients, all mild in nature. Two of our rituximab-treated patients experienced serious AEs requiring surgical interventions: pyoderma gangrenosum vaginalis with perianal abscess and fistula and an increase in the size of a meningioma. No case of progressive multifocal leukoencephalopathy (PML) was detected. Conclusion In our real-world cohort, rituximab was well-tolerated and effective in reducing relapse rate and disability progression in relapsing-remitting and progressive MS patients.
Collapse
|
56
|
Berntsson SG, Kristoffersson A, Boström I, Feresiadou A, Burman J, Landtblom AM. Rapidly increasing off-label use of rituximab in multiple sclerosis in Sweden - Outlier or predecessor? Acta Neurol Scand 2018; 138:327-331. [PMID: 29797711 DOI: 10.1111/ane.12963] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2018] [Indexed: 01/02/2023]
Abstract
OBJECTIVES Off-label use of rituximab to treat MS patients in Sweden is high, and the need for long-term safety data may not be met. Our objectives were to assess the rate of rituximab prescription in patients with multiple sclerosis in Sweden and, in addition, to evaluate the safety of rituximab in a single centre for patients with multiple sclerosis. MATERIAL AND METHODS Review of the Swedish MS register was performed to study the number of MS patients treated with rituximab during the last 6 years. Investigation also included a retrospective review of medical files in search for possible side effects/adverse events in all adult patients with MS treated with rituximab at Uppsala University Hospital. RESULTS Presently, in Sweden the rate of rituximab prescriptions in relation to other annually started of disease- modifying drugs in MS is 53.5%. CONCLUSIONS The share of MS patients in Sweden who are treated with rituximab is very high, and also rapidly increasing. Taken into account the off-label use, cases with adverse medical conditions that could possibly be related to rituximab use should be reported thoroughly.
Collapse
Affiliation(s)
- S. G. Berntsson
- Department of Neuroscience, Neurology; Uppsala University; University Hospital; Uppsala Sweden
| | - A. Kristoffersson
- Department of Neuroscience, Neurology; Uppsala University; University Hospital; Uppsala Sweden
- Neurology Policlinic; Department of Medical Specialist; Motala General Hospital; Motala Sweden
| | - I. Boström
- Department of Clinical and Experimental Medicine, Neurology; Medical Faculty; University of Linköping; Linköping Sweden
| | - A. Feresiadou
- Department of Neuroscience, Neurology; Uppsala University; University Hospital; Uppsala Sweden
| | - J. Burman
- Department of Neuroscience, Neurology; Uppsala University; University Hospital; Uppsala Sweden
| | - A. M. Landtblom
- Department of Neuroscience, Neurology; Uppsala University; University Hospital; Uppsala Sweden
- Department of Clinical and Experimental Medicine, Neurology; Medical Faculty; University of Linköping; Linköping Sweden
- Neurology Policlinic; Department of Medical Specialist; Motala General Hospital; Motala Sweden
| |
Collapse
|
57
|
Windpessl M, Burgstaller S, Kronbichler A, Pieringer H, Kalev O, Karrer A, Wallner M, Thaler J. Progressive Multifocal Leukoencephalopathy Following Combined Rituximab-Based Immune-Chemotherapy for Post-transplant Lymphoproliferative Disorder in a Renal Transplant Recipient: A Case Report. Transplant Proc 2018; 50:881-883. [PMID: 29661457 DOI: 10.1016/j.transproceed.2018.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 01/17/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Transplant recipients are at risk of developing progressive multifocal leukoencephalopathy (PML), an opportunistic infection due to reactivation of JC virus. Post-transplant lymphoproliferative disorders (PTLDs) represent a common malignancy in this population, and antiCD20-therapy has become an established component of its treatment. CASE PRESENTATION We describe the first case of a renal allograft transplant recipient with PTLD who received rituximab-based immune-chemotherapy and developed PML shortly thereafter. Despite early suspicion and diagnosis, the disease ran a relentlessly progressive course, and the patient succumbed to his illness shortly thereafter. CONCLUSION PML should be strongly suspected whenever unusual neurologic symptoms appear in the context of immunosuppression. Clinicians and patients should be aware of the potential for PML after rituximab therapy.
Collapse
Affiliation(s)
- M Windpessl
- Department of Internal Medicine IV, Hematology, Oncology and Nephrology, Academic Teaching Hospital Wels-Grieskirchen, Wels, Austria.
| | - S Burgstaller
- Department of Internal Medicine IV, Hematology, Oncology and Nephrology, Academic Teaching Hospital Wels-Grieskirchen, Wels, Austria
| | - A Kronbichler
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University of Innsbruck, Innsbruck, Austria
| | - H Pieringer
- Academic Research Unit, 2nd Department of Medicine, Kepler University Hospital, Med Campus III, Linz, Austria; Paracelsus Private Medical University, Salzburg, Austria
| | - O Kalev
- Department of Neuropathology, Kepler University Hospital, Neuromed Campus, Linz, Austria
| | - A Karrer
- Department of Radiology, Academic Teaching Hospital Wels-Grieskirchen, Wels, Austria
| | - M Wallner
- Department of Internal Medicine IV, Hematology, Oncology and Nephrology, Academic Teaching Hospital Wels-Grieskirchen, Wels, Austria
| | - J Thaler
- Department of Internal Medicine IV, Hematology, Oncology and Nephrology, Academic Teaching Hospital Wels-Grieskirchen, Wels, Austria
| |
Collapse
|
58
|
Anti-JC virus antibody index changes in rituximab-treated multiple sclerosis patients. J Neurol 2018; 265:2342-2345. [DOI: 10.1007/s00415-018-8996-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/14/2022]
|
59
|
Abstract
In this review article, typical imaging findings of diseases elicited by immunomodulatory drugs are presented. Progressive multifocal leucoencephalopathy (PML), a subacute infection due to reactivation of the JC virus, has a typical magnetic resonance imaging (MRI) pattern, which must be considered in the framework of immunomodulatory multiple sclerosis (MS) treatment with natalizumab, fingolimod, rituximab and fumarate and also in lymphoproliferative diseases or other forms of immunosuppression. Restoration of the immune system may result in an overlapping and overwhelming immune response in terms of an immune reconstitution inflammatory syndrome (IRIS). Further conditions with typical MRI patterns comprise methotrexate-associated leucoencephalopathy and posterior reversible encephalopathy syndrome (PRES). In immunotherapy with activation of T‑lymphocytes, attention should always be paid to the fact that the T‑lymphocytes activated by different processes not only target the diseased tissue but also normal brain tissue.
Collapse
Affiliation(s)
| | - H Urbach
- Klinik für Neuroradiologie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Deutschland.
| |
Collapse
|
60
|
Alcalá C, Gascón F, Pérez-Miralles F, Gil-Perotín S, Navarré A, Boscá I, Coret F, Casanova B. Efficacy and safety of rituximab in relapsing and progressive multiple sclerosis: a hospital-based study. J Neurol 2018; 265:1690-1697. [PMID: 29785523 DOI: 10.1007/s00415-018-8899-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/02/2018] [Accepted: 05/05/2018] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Rituximab is considered as a potential therapeutic option in relapsing-remitting (RRMS) and progressive forms (PMS) of multiple sclerosis (MS). OBJECTIVE To investigate the effectiveness and safety of rituximab in MS. PATIENTS AND METHODS Observational study of effectiveness (clinical and radiological) and safety of rituximab in RRMS and PMS. RESULTS A total of 90 rituximab-treated patients were collected: 31 RRMS and 59 PMS All patients had an active disease despite standard treatment. The annualized relapse rate (ARR) the year before rituximab was 0.86, 53.3% of patients had gadolinium enhanced lesion, and mean Expanded Disability Status Scale (EDSS) had increased from 4.2 to 4.9. During treatment, the ARR was reduced an 88.4% (p < 0.001). A significant decrease of EDSS to 4.6 was observed (p = 0.01) after 1 year of treatment, which remained stable during the second year in both groups. There was no evidence of disease activity in 70% of total sample, 74.2% of RRMS, and 67% of the PMS patients. Infusion-related symptoms were the most prevalent side effect (18.8%) and most were mild. Three thrombotic events were detected. CONCLUSION Rituximab could be an effective and safe treatment in aggressive RRMS. Some selected PMS patients could also benefit from this treatment.
Collapse
Affiliation(s)
- Carmen Alcalá
- Unit of Neuroimmunology, Department of Neurology, Hospital Universitari i Politècnic La Fe, 106 Avenue Fernando Abril Martorell, 46026, Valencia, Spain.
| | - F Gascón
- Unit of Neuroimmunology, Department of Neurology, Hospital Clínico Universitario, Valencia, Spain
| | - F Pérez-Miralles
- Unit of Neuroimmunology, Department of Neurology, Hospital Universitari i Politècnic La Fe, 106 Avenue Fernando Abril Martorell, 46026, Valencia, Spain
| | - S Gil-Perotín
- Unit of Neuroimmunology, Department of Neurology, Hospital Universitari i Politècnic La Fe, 106 Avenue Fernando Abril Martorell, 46026, Valencia, Spain
| | - A Navarré
- Unit of Neuroimmunology, Department of Neurology, Hospital Clínico Universitario, Valencia, Spain
| | - I Boscá
- Unit of Neuroimmunology, Department of Neurology, Hospital Universitari i Politècnic La Fe, 106 Avenue Fernando Abril Martorell, 46026, Valencia, Spain
| | - F Coret
- Unit of Neuroimmunology, Department of Neurology, Hospital Clínico Universitario, Valencia, Spain
| | - B Casanova
- Unit of Neuroimmunology, Department of Neurology, Hospital Universitari i Politècnic La Fe, 106 Avenue Fernando Abril Martorell, 46026, Valencia, Spain
| |
Collapse
|
61
|
Akaishi T, Nakashima I. Efficiency of antibody therapy in demyelinating diseases. Int Immunol 2018; 29:327-335. [PMID: 28910968 DOI: 10.1093/intimm/dxx037] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022] Open
Abstract
Monoclonal antibody therapy is a new treatment strategy for many types of diseases including cancers and autoimmune diseases, realizing a high efficacy and tolerability. In multiple sclerosis (MS) and neuromyelitis optica (NMO) spectrum disorders, several monoclonal antibodies have been suggested to decrease the incidence of clinical relapse and the disease activity. In MS, anti-α4 integrin (natalizumab), anti-CD52 (alemtuzumab), anti-CD25 (daclizumab) and anti-CD20 (ocrelizumab) have been shown to effectively reduce the relapses in randomized controlled trials and have been approved by the Food and Drug Administration. Specifically, ocrelizumab is the first drug that has shown significant suppression of brain volume loss and suppression of chronic disability progression. In NMO, though there have yet to be any approved monoclonal antibodies, rituximab, anti-complement C5 (eculizumab), anti-IL-6 receptor (tocilizumab), anti-CD19 (inebilizumab) and non-pathogenic anti-aquaporin 4 (aquaporumab) have been suggested to be effective, and some of these are now under clinical trials. Aquaporumab is a non-pathogenic recombinant human monoclonal antibody that competitively inhibits the binding of the pathogenic auto-antibody against aquaporin 4 in NMO patients; thus, it is expected to be highly disease specific with less non-specific adverse events. Some of these monoclonal antibodies in MS and NMO are known to cause several notable adverse events. Natalizumab and rituximab increase the risk of progressive multifocal leukoencephalopathy. Eculizumab increases the risk of meningococcal infection. Tocilizumab is known to cause intestinal diverticulitis that can cause intestinal perforation. In this review, we summarize the characteristics of, evidence for and notable adverse events of each monoclonal antibody in MS and NMO.
Collapse
Affiliation(s)
- Tetsuya Akaishi
- Department of Neurology, Tohoku University School of Medicine, Seiryo-machi 1-1, Aoba-ku, Sendai 980-8574, Japan.,Department of Neurology, Yonezawa National Hospital, Misawa 26100-1, Yonezawa 992-1202, Japan
| | - Ichiro Nakashima
- Department of Neurology, Tohoku University School of Medicine, Seiryo-machi 1-1, Aoba-ku, Sendai 980-8574, Japan.,Department of Neurology, Tohoku Medical and Pharmaceutical University, Fukumuro 1-12-1, Miyagino-ku, Sendai 983-8512, Japan
| |
Collapse
|
62
|
Zurawski J, Stankiewicz J. Multiple Sclerosis Re-Examined: Essential and Emerging Clinical Concepts. Am J Med 2018; 131:464-472. [PMID: 29274753 DOI: 10.1016/j.amjmed.2017.11.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/18/2017] [Accepted: 11/21/2017] [Indexed: 11/24/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system characterized by exacerbations of neurological dysfunction due to inflammatory demyelination. Neurologic symptoms typically present in young adulthood and vary based on the site of inflammation, although weakness, sensory impairment, brainstem dysfunction, and vision loss are common. MS occurs more frequently in women and its development is complex-genetics, hormones, geography, vitamin D, and viral exposure all play roles. Early MS is characterized by relapsing-remitting course and inflammation of the white matter, although as patients age, the disease often transitions to a pathologically distinct secondary progressive phase with gradual disability accrual affecting gait, coordination, and bladder function. A minority of patients (10%) have disease that is progressive at onset. In the past decade, there has been a remarkable expansion in disease-modifying therapy for MS, but treatment of progressive disease remains a challenge. This article reviews foundational concepts in MS and emerging work that has reshaped understanding of the disease, providing new insight for therapeutic advance.
Collapse
Affiliation(s)
- Jonathan Zurawski
- Partners MS Center, Boston, Mass; Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - James Stankiewicz
- Partners MS Center, Boston, Mass; Brigham and Women's Hospital, Harvard Medical School, Boston, Mass.
| |
Collapse
|
63
|
Progressive multifocal leukoencephalopathy in rituximab-treated rheumatic diseases: a rare event. J Neurovirol 2018; 24:323-331. [PMID: 29508305 PMCID: PMC5992248 DOI: 10.1007/s13365-018-0615-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 11/19/2022]
Abstract
This report assesses the observed risk of PML in patients treated with the anti-CD20 monoclonal antibody rituximab in the regulatory authority-approved autoimmune indications rheumatoid arthritis (RA), granulomatosis with polyangiitis (GPA), and microscopic polyangiitis (MPA). This was a cumulative analysis of confirmed PML cases in patients receiving rituximab for RA or GPA/MPA from both spontaneous reports and clinical trial sources, as captured in the manufacturer global company safety and clinical databases. Overall reporting rates were calculated and patient case details were summarized. As of 17 November 2015, there were nine confirmed PML cases among patients who had received rituximab for RA and two for GPA. Corresponding estimated reporting rates were 2.56 per 100,000 patients with RA (estimated exposure ≈ 351,396 patients) and < 1 per 10,000 patients with GPA/MPA (estimated exposure 40,000–50,000 patients). In all cases, patients had ≥ 1 potential risk factor for PML independent of rituximab treatment. In the RA population, the estimated reporting rate of PML generally remained stable and low since 2009 despite increasing rituximab exposure. There was no pattern of latency from time of rituximab initiation to PML development and no association of PML with the number of rituximab courses. Global post-marketing safety and clinical trial data demonstrated that the occurrence of PML is very rare among rituximab-treated patients with RA or GPA/MPA and has remained stable over time.
Collapse
|
64
|
Hedrich CM, Smith EMD, Beresford MW. Juvenile-onset systemic lupus erythematosus (jSLE) - Pathophysiological concepts and treatment options. Best Pract Res Clin Rheumatol 2018; 31:488-504. [PMID: 29773269 DOI: 10.1016/j.berh.2018.02.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The systemic autoimmune/inflammatory condition systemic lupus erythematosus (SLE) manifests before the age of 16 years in 10-20% of all cases. Clinical courses are more severe, and organ complications are more common in patients with juvenile SLE. Varying gender distribution in different age groups and increasing severity with younger age and the presence of monogenic disease in early childhood indicate distinct differences in the pathophysiology of juvenile versus adult-onset SLE. Regardless of these differences, classification criteria and treatment options are identical. In this article, we discuss age-specific pathomechanisms of juvenile-onset SLE, which are currently available and as future treatment options, and propose reclassification of different forms of SLE along the inflammatory spectrum from autoinflammation to autoimmunity.
Collapse
Affiliation(s)
- Christian M Hedrich
- Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK.
| | - Eve M D Smith
- Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
| | - Michael W Beresford
- Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
| |
Collapse
|
65
|
Wallin MT. Rituximab is an acceptable alternative to ocrelizumab for treating multiple sclerosis – No. Mult Scler 2018; 24:1159-1161. [DOI: 10.1177/1352458518757931] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Mitchell T Wallin
- VA Multiple Sclerosis Center of Excellence-East, Washington, DC, USA
- School of Medicine, Georgetown University, Washington, DC, USA
| |
Collapse
|
66
|
T cell deficiencies as a common risk factor for drug associated progressive multifocal leukoencephalopathy. Immunobiology 2018; 223:508-517. [PMID: 29472141 DOI: 10.1016/j.imbio.2018.01.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/19/2018] [Accepted: 01/21/2018] [Indexed: 01/07/2023]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a disease of the central nervous system caused by neuropathogenic prototypes of ubiquitous community-acquired JC virus (JCV). The disease became of particular concern following its association with certain therapies that modulate immune system function without heavy immunosuppression. Due to lack of prophylactic/treatment options and poor outcomes, which often include severe disability or death, PML is a considerable concern for development of new drugs that interfere with immune system functions. In this review of clinical and research findings, we discuss the evidence that deficiencies in CD4+ T helper cells, cytotoxic CD8+ T cells, and interferon gamma are of crucial importance for development of PML under a variety of circumstances, including those associated with use of various drugs, regardless of differences in their mechanisms of action. These deficiencies apparently enable transformation of the harmless JCV archetype into neuropathogenic prototypes, but the site(s), and the mechanisms, of this transformation are yet to be elucidated. Here we discuss the evidence for brain as one of the sites of this transformation, and propose a model of PML pathogenesis that emphasizes the central role of T cell deficiencies in the two life cycles of the JCV, one non-pathogenic and one neuropathogenic. Finally, we conclude that the development of clinical grade T cell functional tests and more consistent use of already available laboratory tests for T cell subset analysis would greatly aid the effort to more accurately predict and assess the magnitude of PML risk for concerned therapeutic interventions.
Collapse
|
67
|
Sharif K, Watad A, Bragazzi NL, Asher E, Abu Much A, Horowitz Y, Lidar M, Shoenfeld Y, Amital H. Anterior ST-elevation myocardial infarction induced by rituximab infusion: A case report and review of the literature. J Clin Pharm Ther 2018; 42:356-362. [PMID: 28440561 DOI: 10.1111/jcpt.12522] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/05/2017] [Indexed: 12/23/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVES Rituximab is a chimeric monoclonal anti-CD20 antibody approved for the treatment of some lymphoid malignancies as well as for autoimmune diseases including rheumatoid arthritis (RA), idiopathic thrombocytopenic purpura (ITP) and vasculitis. Generally, rituximab is well tolerated; nevertheless, some patients develop adverse effects including infusion reactions. Albeit rare, these reactions may in some cases be life-threatening conditions. Rituximab cardiovascular side effects include more common effects such as hypertension, oedema and rare cases of arrhythmias and myocardial infarction. CASE SUMMARY In this article, we report a case of a 58-year-old man with a history of overlap syndrome including RA and limited scleroderma who was treated with rituximab and developed a dramatic ST-elevation myocardial infarction (STEMI) during the drug administration. WHAT IS NEW AND CONCLUSION This report underlines previous published reports emphasizing the awareness of such an association. This communication also warrants the importance of screening for ischaemic heart disease in selected cases of patients treated with rituximab.
Collapse
Affiliation(s)
- K Sharif
- Department of Medicine "B", Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel
| | - A Watad
- Department of Medicine "B", Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - N L Bragazzi
- School of Public Health, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - E Asher
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,The Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - A Abu Much
- Department of Medicine "B", Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,The Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Y Horowitz
- Department of Medicine "B", Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel
| | - M Lidar
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,Rheumatology Unit, Sheba Medical Center, Tel-Hashomer, Israel
| | - Y Shoenfeld
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - H Amital
- Department of Medicine "B", Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|
68
|
Memon AB, Javed A, Caon C, Srivastawa S, Bao F, Bernitsas E, Chorostecki J, Tselis A, Seraji-Bozorgzad N, Khan O. Long-term safety of rituximab induced peripheral B-cell depletion in autoimmune neurological diseases. PLoS One 2018; 13:e0190425. [PMID: 29309416 PMCID: PMC5757948 DOI: 10.1371/journal.pone.0190425] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 12/14/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND B-cells play a pivotal role in several autoimmune diseases, including patients with immune-mediated neurological disorders (PIMND), such as neuromyelitis optica (NMO), multiple sclerosis (MS), and myasthenia gravis (MG). Targeting B-cells has been an effective approach in ameliorating both central and peripheral autoimmune diseases. However, there is a paucity of literature on the safety of continuous B-cell depletion over a long period of time. OBJECTIVE The aim of this study was to examine the long-term safety, incidence of infections, and malignancies in subjects receiving continuous therapy with a B-cell depleting agent rituximab over at least 3 years or longer. METHODS This was a retrospective study involving PIMND who received continuous cycles of rituximab infusions every 6 to 9 months for up to 7 years. The incidence of infection related adverse events (AE), serious adverse events (SAE), and malignancies were observed. RESULTS There were a total of 32 AE and 4 SAE with rituximab treatment. The 3 SAE were noted after 9 cycles (48 months) and 1 SAE was observed after 11 cycles (60 months) of rituximab. There were no cases of Progressive multifocal leukoencephalopathy (PML) and malignancies observed throughout the treatment period. Rituximab was well tolerated without any serious infusion reactions. Also, rituximab was found to be beneficial in treating PIMND over a 7-year period. CONCLUSIONS This study demonstrates that long-term depletion of peripheral B-cells appears safe and efficacious in treating PIMND. Longer and larger prospective studies with rituximab are needed to carefully ascertain risks associated with chronic B-cell depletion, including malignancies. Recognizing that this is a small, retrospective study, such data nonetheless complement the growing literature documenting the safety and tolerability of B-cell depleting agents in neurological diseases.
Collapse
Affiliation(s)
- Anza B Memon
- Multiple Sclerosis Center, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Adil Javed
- Multiple Sclerosis Center, Department of Neurology, University of Chicago Pritzker School of Medicine, Chicago, Illinois, United States of America
| | - Christina Caon
- Multiple Sclerosis Center, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Shitiz Srivastawa
- Multiple Sclerosis Center, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Fen Bao
- The Sastry Foundation Advanced Imaging Laboratory, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Evanthia Bernitsas
- Multiple Sclerosis Center, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Jessica Chorostecki
- Multiple Sclerosis Center, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America.,The Sastry Foundation Advanced Imaging Laboratory, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Alexandros Tselis
- Multiple Sclerosis Center, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Navid Seraji-Bozorgzad
- The Sastry Foundation Advanced Imaging Laboratory, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Omar Khan
- Multiple Sclerosis Center, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America.,The Sastry Foundation Advanced Imaging Laboratory, Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| |
Collapse
|
69
|
Molloy ES, Calabrese CM, Calabrese LH. The Risk of Progressive Multifocal Leukoencephalopathy in the Biologic Era: Prevention and Management. Rheum Dis Clin North Am 2017; 43:95-109. [PMID: 27890176 DOI: 10.1016/j.rdc.2016.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a rare, typically fatal, demyelinating central nervous system infection caused by reactivation of the John Cunningham virus that generally occurs in immunosuppressed patients. With an evolving understanding of a greater clinical heterogeneity of PML and significant implications for therapy, PML should be considered in the differential diagnosis of neurologic presentations of rheumatic diseases. Increased awareness of PML among rheumatologists is required, as earlier diagnosis and restoration of immune function may improve the otherwise grim prognosis associated with PML.
Collapse
Affiliation(s)
- Eamonn S Molloy
- Department of Rheumatology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.
| | - Cassandra M Calabrese
- RJ Fasenmeyer Center for Clinical Immunology, Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Leonard H Calabrese
- RJ Fasenmeyer Center for Clinical Immunology, Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
70
|
Gelfand JM, Cree BAC, Hauser SL. Ocrelizumab and Other CD20 + B-Cell-Depleting Therapies in Multiple Sclerosis. Neurotherapeutics 2017; 14:835-841. [PMID: 28695471 PMCID: PMC5722762 DOI: 10.1007/s13311-017-0557-4] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Selective depletion of CD20+ B cells by anti-CD20 monoclonal antibodies as monotherapy in multiple sclerosis (MS) profoundly suppresses acute inflammatory disease activity and signifies an important advance in the treatment of relapsing-remitting MS. Ocrelizumab, a humanized anti-CD20 monoclonal antibody, is also the first proven therapy to lessen disability progression in primary progressive MS-a breakthrough for patients with a disease that had no proven therapy. Ocrelizumab is generally well tolerated, with the most common adverse events experienced being infusion reactions and infections. In ocrelizumab trials in MS a numerical imbalance in the risk of malignancies was observed. In this article, we review advances in anti-CD20 B-cell-depleting biological therapies for MS, including ocrelizumab, rituximab, and ofatumumab.
Collapse
Affiliation(s)
- Jeffrey M Gelfand
- Multiple Sclerosis Center, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Bruce A C Cree
- Multiple Sclerosis Center, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Stephen L Hauser
- Multiple Sclerosis Center, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA.
| |
Collapse
|
71
|
Grebenciucova E, Pruitt A. Infections in Patients Receiving Multiple Sclerosis Disease-Modifying Therapies. Curr Neurol Neurosci Rep 2017; 17:88. [PMID: 28940162 DOI: 10.1007/s11910-017-0800-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW This paper will systemically review the risk of infections associated with current disease-modifying treatments and will discuss pre-treatment testing recommendations, infection monitoring strategies, and patient education. RECENT FINDINGS Aside from glatiramer acetate and interferon-beta therapies, all other multiple sclerosis treatments to various degrees impair immune surveillance and may predispose patients to the development of both community-acquired and opportunistic infections. Some of these infections are rarely seen in neurologic practice, and neurologists should be aware of how to monitor for these infections and how to educate patients about medication-specific risks. Of particular interest in this discussion is the risk of PML in association with the recently approved B cell depleting therapy, ocrelizumab, particularly when switching from natalizumab. The risk of infection in association with MS treatments has become one of the most important factors in the choice of therapy. Balance of the overall risk versus benefit should be continuously re-evaluated during treatment.
Collapse
Affiliation(s)
- Elena Grebenciucova
- Multiple Sclerosis Division, Davee Department of Neurology and Clinical Neurosciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Amy Pruitt
- Multiple Sclerosis Division, the Department of Neurology, Perelman School of Medicine, The University of Pennsylvania, 3400 Convention Avenue, Philadelphia, PA, 19104, USA
| |
Collapse
|
72
|
Bradshaw MJ, Cho TA, Chow FC. Central Nervous System Infections Associated with Immunosuppressive Therapy for Rheumatic Disease. Rheum Dis Clin North Am 2017; 43:607-619. [PMID: 29061246 DOI: 10.1016/j.rdc.2017.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Patients on immunosuppressive therapy for rheumatic diseases are at increased risk of infection. Although infections of the central nervous system (CNS) are less common compared with other sites, patients on broadly immunosuppressive and biologic immunomodulatory agents may be susceptible to more severe, disseminated forms of infection, including of the CNS. Certain key principles regarding infection risk apply across immunosuppressive therapies, including increased risk with higher doses and longer duration of therapy and with combination therapy. Providers should be aware of the CNS infection risk related to immunosuppressant use to help guide best practices for screening and prophylaxis.
Collapse
Affiliation(s)
- Michael J Bradshaw
- Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Massachusetts General Hospital, 60 Fenwood Road, 4th floor, Boston, MA 02115, USA
| | - Tracey A Cho
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Felicia C Chow
- Department of Neurology and Division of Infectious Diseases, University of California, San Francisco, Zuckerberg San Francisco General Hospital, 1001 Potrero Avenue, Building 1, Room 101, San Francisco, CA 94110, USA.
| |
Collapse
|
73
|
Pathophysiology of thrombotic thrombocytopenic purpura. Blood 2017; 130:1181-1188. [PMID: 28768626 DOI: 10.1182/blood-2017-04-636431] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/27/2017] [Indexed: 01/29/2023] Open
Abstract
The discovery of a disintegrin-like and metalloproteinase with thrombospondin type 1 motif, member 13 (ADAMTS13) revolutionized our approach to thrombotic thrombocytopenic purpura (TTP). Inherited or acquired ADAMTS13 deficiency allows the unrestrained growth of microthrombi that are composed of von Willebrand factor and platelets, which account for the thrombocytopenia, hemolytic anemia, schistocytes, and tissue injury that characterize TTP. Most patients with acquired TTP respond to a combination of plasma exchange and rituximab, but some die or acquire irreversible neurological deficits before they can respond, and relapses can occur unpredictably. However, knowledge of the pathophysiology of TTP has inspired new ways to prevent early deaths by targeting autoantibody production, replenishing ADAMTS13, and blocking microvascular thrombosis despite persistent ADAMTS13 deficiency. In addition, monitoring ADAMTS13 has the potential to identify patients who are at risk of relapse in time for preventive therapy.
Collapse
|
74
|
Louthrenoo W, Kasitanon N, Katchamart W, Aiewruengsurat D, Chevaisrakul P, Chiowchanwisawakit P, Dechanuwong P, Hanvivadhanakul P, Mahakkanukrauh A, Manavathongchai S, Muangchan C, Narongroeknawin P, Phumethum V, Siripaitoon B, Suesuwan A, Suwannaroj S, Uea-Areewongsa P, Ukritchon S, Asavatanabodee P, Koolvisoot A, Nanagara R, Totemchokchyakarn K, Nuntirooj K, Kitumnuaypong T. 2016 updated Thai Rheumatism Association Recommendations for the use of biologic and targeted synthetic disease-modifying anti-rheumatic drugs in patients with rheumatoid arthritis. Int J Rheum Dis 2017; 20:1166-1184. [DOI: 10.1111/1756-185x.13130] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Worawit Louthrenoo
- Division of Rheumatology; Department of Internal Medicine; Faculty of Medicine; Chiang Mai University; Chiang Mai Thailand
| | - Nuntana Kasitanon
- Division of Rheumatology; Department of Internal Medicine; Faculty of Medicine; Chiang Mai University; Chiang Mai Thailand
| | - Wanruchada Katchamart
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Siriraj Hospital, Mahidol University; Bangkok Thailand
| | - Duangkamol Aiewruengsurat
- Division of Rheumatology; Department of Internal Medicine; Faculty of Medicine; Prince of Songkla University; Songkla Thailand
| | - Parawee Chevaisrakul
- Division of Allergy Immunology and Rheumatology; Department of Internal Medicine; Faculty of Medicine; Ramathibodi Hospital, Mahidol University; Bangkok Thailand
| | - Praveena Chiowchanwisawakit
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Siriraj Hospital, Mahidol University; Bangkok Thailand
| | - Pornchai Dechanuwong
- Department of Internal Medicine; Faculty of Medicine; Vajira Hospital, Navamindradhiraj University; Bangkok Thailand
| | - Punchong Hanvivadhanakul
- Division of Rheumatology; Department of Internal Medicine; Faculty of Medicine; Thammasat University; Pathum Thani Thailand
| | - Ajanee Mahakkanukrauh
- Division of Allergy Immunology and Rheumatology; Department of Medicine, Faculty of Medicine; Khon Kaen University; Khon Kaen Thailand
| | - Siriporn Manavathongchai
- Department of Internal Medicine; Faculty of Medicine; Vajira Hospital, Navamindradhiraj University; Bangkok Thailand
| | - Chayawee Muangchan
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Siriraj Hospital, Mahidol University; Bangkok Thailand
| | - Pongthorn Narongroeknawin
- Division of Rheumatology, Department of Internal Medicine; Phramongkutklao Hospital and College of Medicine; Bangkok Thailand
| | - Veerapong Phumethum
- Division of Rheumatology, Department of Internal Medicine; Pha Pok Klao Hospital; Chanthaburi Thailand
| | - Boonjing Siripaitoon
- Division of Rheumatology; Department of Internal Medicine; Faculty of Medicine; Prince of Songkla University; Songkla Thailand
| | | | - Siraphop Suwannaroj
- Division of Allergy Immunology and Rheumatology; Department of Medicine, Faculty of Medicine; Khon Kaen University; Khon Kaen Thailand
| | - Parichat Uea-Areewongsa
- Division of Rheumatology; Department of Internal Medicine; Faculty of Medicine; Prince of Songkla University; Songkla Thailand
| | - Sittichai Ukritchon
- Division of Rheumatology, Department of Medicine; Faculty of Medicine; Chulalongkorn University; Bangkok Thailand
| | - Paijit Asavatanabodee
- Rheumatic Disease Unit; Department of Medicine; Phramongkutklao Hospital; Bangkok Thailand
| | - Ajchara Koolvisoot
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Siriraj Hospital, Mahidol University; Bangkok Thailand
| | - Ratanavadee Nanagara
- Division of Allergy Immunology and Rheumatology; Department of Medicine, Faculty of Medicine; Khon Kaen University; Khon Kaen Thailand
| | - Kitti Totemchokchyakarn
- Division of Allergy Immunology and Rheumatology; Department of Internal Medicine; Faculty of Medicine; Ramathibodi Hospital, Mahidol University; Bangkok Thailand
| | - Kanokrut Nuntirooj
- Division of Allergy Immunology and Rheumatology; Department of Internal Medicine; Faculty of Medicine; Ramathibodi Hospital, Mahidol University; Bangkok Thailand
| | - Tasanee Kitumnuaypong
- Rheumatology Unit; Department of Medicine; Rajavithi Hospital, Ministry of Public Health; Bangkok Thailand
| |
Collapse
|
75
|
Mornese Pinna S, Scarvaglieri E, Milia MG, Imperiale D, Ghisetti V, Audagnotto S, Prochet A, Lipani F, Bonora S, Di Perri G, Calcagno A. Detectable cerebrospinal fluid JCV DNA in late-presenting HIV-positive patients: beyond progressive multifocal leukoencephalopathy? J Neurovirol 2017; 23:763-767. [PMID: 28681343 DOI: 10.1007/s13365-017-0549-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/08/2017] [Accepted: 06/20/2017] [Indexed: 10/19/2022]
Abstract
In the absence of effective prophylaxis and treatment, therapeutic options in HIV-positive patients with progressive multifocal leukoencephalopathy (PML) are limited to antiretroviral therapy: nevertheless, outcome is poor. We conducted a retrospective study (2009-2015) describing the outcome of 25 HIV-positive patients with detectable cerebrospinal fluid JC virus DNA: 14 had a probable PML while the others had evidence of other inflammatory central nervous system (CNS) affecting disorders. In the former group, 6-month mortality was 45.5% vs 21.4 in the latter one: survival was higher than previously described but no predictor of poor outcome was identified. Two patients treated with 5HT2-inhibitors survived. The contributing role of JCV replication in other CNS-affecting disorders needs to be assessed as well as the benefits of 5HT2-inhibitors in HIV-positive patients with proven PML.
Collapse
Affiliation(s)
- S Mornese Pinna
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Amedeo di Savoia Hospital, C.so Svizzera 164, 10149, Turin, Italy.
| | - E Scarvaglieri
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Amedeo di Savoia Hospital, C.so Svizzera 164, 10149, Turin, Italy
| | - M G Milia
- Laboratory of Microbiology and Molecular Biology, Ospedale Amedeo di Savoia, ASL TO2, Turin, Italy
| | - D Imperiale
- Unit of Neurology, Ospedale Maria Vittoria, ASL TO2, Turin, Italy
| | - V Ghisetti
- Laboratory of Microbiology and Molecular Biology, Ospedale Amedeo di Savoia, ASL TO2, Turin, Italy
| | - S Audagnotto
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Amedeo di Savoia Hospital, C.so Svizzera 164, 10149, Turin, Italy
| | - A Prochet
- Unit of Radiology, Ospedale San Giovanni Bosco, ASL TO2, Turin, Italy
| | - F Lipani
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Amedeo di Savoia Hospital, C.so Svizzera 164, 10149, Turin, Italy
| | - S Bonora
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Amedeo di Savoia Hospital, C.so Svizzera 164, 10149, Turin, Italy
| | - G Di Perri
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Amedeo di Savoia Hospital, C.so Svizzera 164, 10149, Turin, Italy
| | - A Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Amedeo di Savoia Hospital, C.so Svizzera 164, 10149, Turin, Italy
| |
Collapse
|
76
|
Abstract
In patients with membranous nephropathy, alkylating agents (cyclophosphamide or chlorambucil) alone or in combination with steroids achieve remission of nephrotic syndrome more effectively than conservative treatment or steroids alone, but can cause myelotoxicity, infections, and cancer. Calcineurin inhibitors can improve proteinuria, but are nephrotoxic. Most patients relapse after treatment withdrawal and can become treatment dependent, which increases the risk of nephrotoxicity. The discovery of nephritogenic autoantibodies against podocyte M-type phospholipase A2 receptor (PLA2R) and thrombospondin type-1 domain- containing protein 7A (THSD7A) antigens provides a clear pathophysiological rationale for interventions that specifically target B-cell lineages to prevent antibody production and subepithelial deposition. The anti-CD20 monoclonal antibody rituximab is safe and achieves remission of proteinuria in approximately two-thirds of patients with membranous nephropathy. In those with PLA2R-related disease, remission can be predicted by anti-PLA2R antibody depletion and relapse by antibody re-emergence into the circulation. Thus, integrated evaluation of serology and proteinuria could guide identification of affected patients and treatment with individually tailored protocols. Nonspecific and toxic immunosuppressive regimens will fall out of use. B-cell modulation by rituximab and second-generation anti-CD20 antibodies (or plasma cell-targeted therapy in anti-CD20 resistant forms of disease) will lead to a novel therapeutic paradigm for patients with membranous nephropathy.
Collapse
|
77
|
Xie Q, Li X, Sun J, Yuan B, Li Y, Wang L, Wang M. A meta-analysis to determine the efficacy and tolerability of anti-B-cell monoclonal antibodies in multiple sclerosis. Exp Ther Med 2017; 13:3061-3066. [PMID: 28587380 DOI: 10.3892/etm.2017.4298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 12/19/2016] [Indexed: 11/06/2022] Open
Abstract
The present study performed a meta-analysis of randomized controlled trials in multiple sclerosis (MS) patients to evaluate the efficacy and safety of anti-B-cell monoclonal antibodies (mAbs). To the best of our knowledge, no previous meta-analysis has evaluated this. Relevant studies published until March 2015 were retrieved from the PubMed, EMBASE and Cochrane Library using the following keywords: 'Clinical trial', 'randomized', 'multiple sclerosis' or 'MS' and 'monoclonal antibodies' or 'mAbs'. Two authors independently selected the articles and extracted the data. The meta-analysis was performed using Review Manager version 5.3 software. Four randomized clinical trials comprising a total of 745 patients were selected. Anti-B-cell mAb treatment reduced the formation of gadolinium-enhancing lesions [mean difference (MD)=-5.62; 95% confidence interval (CI)=-8.00 to -3.24; P<0.001) and was associated with smaller volume changes of lesions on T2-weighted magnetic resonance imaging (MD=-604.40; 95% CI=-941.23 to -267.57; P<0.001). It also significantly reduced the proportion of MS patients having at least one relapse [odds ratio (OR)=0.25; 95% CI=0.14-0.44; P<0.001). Compared to placebo, anti-B-cell mAb treatment did not increase the frequency of adverse events (OR=0.90; 95% CI=0.54-1.49; P=0.68) and serious adverse events (OR=1.13; 95% CI=0.70-1.80; P=0.62). In conclusion, the present meta-analysis suggested that anti-B-cell mAbs are a relatively effective and safe treatment for MS.
Collapse
Affiliation(s)
- Qinfang Xie
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Xiaoling Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Jingjie Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Boyao Yuan
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Yijun Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Lijuan Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
78
|
Misbah SA. Progressive multi-focal leucoencephalopathy - driven from rarity to clinical mainstream by iatrogenic immunodeficiency. Clin Exp Immunol 2017; 188:342-352. [PMID: 28245526 PMCID: PMC5422720 DOI: 10.1111/cei.12948] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2017] [Indexed: 12/21/2022] Open
Abstract
Advances in immune-mediated targeted therapies have proved to be a double-edged sword for patients by highlighting the risk of iatrogenic infective complications. This has been exemplified by progressive multi-focal leucoencephalopathy (PML), a hitherto rare devastating viral infection of the brain caused by the neurotrophic JC polyoma virus. While PML achieved prominence during the first two decades of the HIV epidemic, effective anti-retroviral treatment and restitution of T cell function has led to PML being less prominent in this population. HIV infection as a predisposing factor has now been supplanted by T cell immunodeficiency induced by a range of immune-mediated therapies as a major cause of PML. This review focuses on PML in the context of therapeutic immunosuppression and encompasses therapeutic monoclonal antibodies, novel immunomodulatory agents such as Fingolimod and dimethyl fumarate, as well as emerging data on PML in primary immune deficiency.
Collapse
Affiliation(s)
- S A Misbah
- Department of Clinical Immunology, Oxford University Hospitals, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
79
|
Clavel G, Moulignier A, Semerano L. Progressive multifocal leukoencephalopathy and rheumatoid arthritis treatments. Joint Bone Spine 2017; 84:671-675. [PMID: 28323224 DOI: 10.1016/j.jbspin.2017.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2016] [Indexed: 12/15/2022]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease of the central nervous system due to reactivation of the JC virus (JCV). PML is extremely uncommon despite the high prevalence of the virus in the general population. No specific treatment is available, and the prognosis is bleak. The diagnosis is based on brain imaging findings, detection of the JCV genome in cerebrospinal fluid samples and, in some cases, histological studies of the brain lesions. The pathophysiological mechanisms that drive the development of PML are incompletely understood. However, a consistent feature is the presence of a predisposing factor, most notably immunosuppression. The risk of developing PML varies with the underlying disease (e.g., HIV infection or autoimmune disease) and with the drugs used to treat them. Biologics have been ranked according to the risk of PML during their use. Natalizumab, a monoclonal antibody given to treat multiple sclerosis, is among the drugs associated with a high risk of PML. Patients given natalizumab are now closely monitored based on anti-JCV antibody titers and index values. In rheumatology, the expanding use of biologics has led to an increase in cases of PML, with rituximab being associated with the highest risk. Given the absence of specific recommendations, exhaustive registries and postmarketing observational studies are urgently needed to gauge the risk of PML according to the underlying disease and drug treatments, with the goal of defining optimal monitoring protocols.
Collapse
Affiliation(s)
- Gaëlle Clavel
- Service de médecine interne, Fondation A. de Rothschild, 25-29, rue Manin, 75019 Paris, France; Inserm UMR 1125, 74, rue Marcel-Cachin, 93017 Bobigny, France; Sorbonne Paris Cité, université Paris 13, 74, rue Marcel-Cachin, 93017 Bobigny, France.
| | | | - Luca Semerano
- Inserm UMR 1125, 74, rue Marcel-Cachin, 93017 Bobigny, France; Sorbonne Paris Cité, université Paris 13, 74, rue Marcel-Cachin, 93017 Bobigny, France; Service de rhumatologie, groupe hospitalier Avicenne Jean-Verdier-René-Muret, Assistance publique-Hopitaux de Paris (AP-HP), 93017 Bobigny, France
| |
Collapse
|
80
|
What's new in neuromyelitis optica? A short review for the clinical neurologist. J Neurol 2017; 264:2330-2344. [PMID: 28289845 DOI: 10.1007/s00415-017-8445-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 02/27/2017] [Indexed: 02/06/2023]
Abstract
The evolution of neuromyelitis optica spectrum disorder (NMOSD) from a rare, incurable and misunderstood disease with almost universally poor outcomes to its present state in just over a decade is unprecedented in neurology and possibly in medicine. Our knowledge of NMOSD biology has led to the recognition of wider phenotypes, new disease mechanisms, and thus clinical trials of new and effective treatments. This article aims to update readers on the recent developments in NMOSD with particular emphasis on clinical advances, the 2015 diagnostic criteria, biomarkers, imaging, and therapeutic interventions.
Collapse
|
81
|
|
82
|
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system most often characterized by clinical relapses and periods of remission. RECENT FINDINGS The past decade has seen a dramatic increase in disease-modifying therapies for MS. Fourteen FDA-approved immunomodulatory drugs are currently available, and more medications are in development. A growing number of reported opportunistic infections, including progressive multifocal leukoencephalopathy (PML), highlight the serious complications of these new drugs and the need for specific screening guidelines. Using data from Phase II and III randomized controlled trials, case reports, drug manufacturing data, and clinical experience, we outline the most common and serious infections associated with novel MS therapies.
Collapse
|
83
|
Stieglbauer K, Pichler R, Topakian R. 10-year-outcomes after rituximab for myasthenia gravis: Efficacy, safety, costs of inhospital care, and impact on childbearing potential. J Neurol Sci 2017; 375:241-244. [PMID: 28320139 DOI: 10.1016/j.jns.2017.02.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/15/2017] [Accepted: 02/03/2017] [Indexed: 12/12/2022]
Abstract
Rituximab (RTX) has emerged as an attractive off-label treatment option for patients with myasthenia gravis (MG) refractory to other immune therapies. However, data on long-term outcome after RTX for MG are still scarce. Here we present the 10-year outcomes [median (range) 10.1 (6.7-11.2) years] with respect to efficacy, safety, costs of inhospital care, and impact on childbearing potential in all four MG patients treated by one of the authors with RTX. In all patients, RTX led to sustained clinical improvement and eventual tapering of other immune therapies. RTX was well tolerated, and complications were not observed. After the start of RTX, annual costs for hospital admissions were markedly reduced compared to costs in the year preceding RTX. Under close clinical observation, two patients had uncomplicated pregnancies giving birth to a healthy child. With regard to its efficacy, excellent tolerance, lack of complications, low frequency of repeat infusions and pending patent expiry in many countries, RTX appears to compare favourably with other immune therapies used for MG. Multicentre trials and registries are urgently needed to further address long-term safety issues and clarify the efficacy and role of RTX in managing MG.
Collapse
Affiliation(s)
| | - Robert Pichler
- Institute of Nuclear Medicine, Neuromed Campus, Johannes Kepler University, Linz, Austria.
| | - Raffi Topakian
- Department of Neurology, Academic Teaching Hospital Wels-Grieskirchen, Wels, Austria.
| |
Collapse
|
84
|
Feng JJ, Ontaneda D. Treating primary-progressive multiple sclerosis: potential of ocrelizumab and review of B-cell therapies. Degener Neurol Neuromuscul Dis 2017; 7:31-45. [PMID: 30050376 PMCID: PMC6053100 DOI: 10.2147/dnnd.s100096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multiple sclerosis (MS) therapy has evolved rapidly with an increased availability of several immunomodulating therapies over the past two decades. Disease-modifying therapies have proven to be effective in treating relapse-remitting MS (RRMS). However, clinical trials involving some of the same agents for secondary-progressive and primary-progressive MS (SPMS and PPMS) have been largely negative. The pathogenesis of progressive MS remains unclear, but B-cells may play a significant role in chronic compartmentalized inflammation, likely contributing to disease progression. Biologics targeted at B-cells, such as rituximab, are effective in treating RRMS. Ocrelizumab is a humanized monoclonal antibody to CD20+ B-cells that has shown positive results in PPMS with a significant reduction in disease progression. This review aims to discuss in detail the involvement of B-cells in MS pathogenesis, current progress of currently available and investigational biologics, with focus on ocrelizumab, and future prospects for B-cell therapy in PPMS.
Collapse
Affiliation(s)
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, OH, USA,
| |
Collapse
|
85
|
Pham TA, Lee JI, Melcher ML. Kidney paired exchange and desensitization: Strategies to transplant the difficult to match kidney patients with living donors. Transplant Rev (Orlando) 2017; 31:29-34. [PMID: 28284304 DOI: 10.1016/j.trre.2017.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 01/06/2017] [Indexed: 12/14/2022]
Abstract
With organs in short supply, only a limited number of kidney transplants can be performed a year. Live donor donation accounts for 1/3rd of all kidney transplants performed in the United States. Unfortunately, not every donor recipient pair is feasible because of Human leukocyte antigen (HLA) sensitization and ABO incompatibility. To overcome these barriers to transplant, strategies such as kidney paired donation (KPD) and desensitization have been developed. KPD is the exchange of donors between at least two incompatible donor-recipient pairs such that they are now compatible. Desensitization is the removal of circulating donor specific antibodies to prevent graft rejection. Regardless of the treatment strategy, highly sensitized patients whose calculated panel reactive antibody (cPRA) is ≥95% remain difficult to transplant with match rates as low as 15% in KPD pools. Desensitization has proved to be difficult in those with high antibody titers. A novel approach is the combination of both KPD and desensitization to facilitate compatible and successful transplantation. A highly sensitized patient can be paired with a better immunological match in the KPD pool and subsequently desensitized to a lesser degree. This article reviews the current progress in KPD and desensitization and their use as a combined therapy.
Collapse
Affiliation(s)
- Thomas A Pham
- Department of Surgery, Division of Hepatobiliary Surgery and Abdominal Transplantation, Keck School of Medicine at University of Southern California, 1510 San Pablo St Suite 514, Los Angeles, CA 90033.
| | - Jacqueline I Lee
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, 750 Welch Ave Suite 200, Palo Alto, CA 94304.
| | - Marc L Melcher
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, 750 Welch Ave Suite 200, Palo Alto, CA 94304.
| |
Collapse
|
86
|
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease that leads to destruction of pancreatic β cells, lifelong dependence on insulin, and increased morbidity and mortality from diabetes-related complications. Preservation of residual β cells at diagnosis is a major goal because higher levels of endogenous insulin secretion are associated with better short- and long-term outcomes. For the past 3 decades, a variety of immune interventions have been evaluated in the setting of new-onset T1D, including nonspecific immunosuppression, pathway-specific immune modulation, antigen-specific therapies, and cellular therapies. To date, no single intervention has produced durable remission off therapy in most treated patients, but the field has gained valuable insights into disease mechanisms and potential immunologic correlates of success. In particular, T-cell–directed therapies, including therapies that lead to partial depletion or modulation of effector T cells and preservation or augmentation of regulatory T cells, have shown the most success and will likely form the backbone of future approaches. The next phase will see evaluation of rational combinations, comprising one or more of the following: an effector T-depleting or -modulating drug, a cytokine-based tolerogenic (regulatory T-cells–promoting) agent, and an antigen-specific component. The long term goal is to reestablish immunologic tolerance to β cells, thereby preserving residual β cells early after diagnosis or enabling restoration of β-cell mass from autologous stem cells or induced neogenesis in patients with established T1D.
Collapse
|
87
|
Infections Associated with Immunobiologics. Infect Dis (Lond) 2017. [DOI: 10.1016/b978-0-7020-6285-8.00088-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
88
|
Humphreys J, Hyrich K, Symmons D. What is the impact of biologic therapies on common co-morbidities in patients with rheumatoid arthritis? Arthritis Res Ther 2016; 18:282. [PMID: 27906042 PMCID: PMC5134078 DOI: 10.1186/s13075-016-1176-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Biologic therapies have revolutionised disease control in patients with rheumatoid arthritis (RA). Theoretically, they have the potential to influence co-morbid disease associated with RA through better control of systemic inflammation. Conversely, co-morbidity may occur as an adverse effect of the drugs. The latest evidence from observational data shows an increased risk of infection in the first 6 months of treatment with tumour necrosis factor inhibitor (TNFi) therapies and potentially other biologic therapies. Rates of infection after the first 6 months decrease and become comparable to patients with RA treated with conventional synthetic disease modifying anti-rheumatic drugs (csDMARDs). TNFi also appear to reduce the risk of cardiovascular disease in these patients, in particular ischaemic heart disease. TNFi treatment may be associated with a small increase in the risk of developing squamous cell carcinoma of the skin; in terms of other cancers, rates appears to be no different to those seen in patients treated with csDMARDs. There is a paucity of data on the impact of other biologic therapies and the effect of all biologic therapies on other common co-morbidities.
Collapse
Affiliation(s)
- Jenny Humphreys
- Arthritis Research UK Centre for Epidemiology, University of Manchester, Manchester, UK
| | - Kimme Hyrich
- Arthritis Research UK Centre for Epidemiology, University of Manchester, Manchester, UK.,NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Deborah Symmons
- Arthritis Research UK Centre for Epidemiology, University of Manchester, Manchester, UK. .,NIHR Manchester Musculoskeletal Biomedical Research Unit, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK. .,Arthritis Research UK Centre for Epidemiology, Manchester Academic Health Science Centre, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
89
|
Mota P, Reddy V, Isenberg D. Improving B-cell depletion in systemic lupus erythematosus and rheumatoid arthritis. Expert Rev Clin Immunol 2016; 13:667-676. [PMID: 27841031 DOI: 10.1080/1744666x.2017.1259068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Rituximab-based B-cell depletion (BCD) therapy is effective in refractory rheumatoid arthritis (RA) and although used to treat patients with refractory systemic lupus erythematosus (SLE) in routine clinical practice, rituximab failed to meet the primary endpoints in two large randomised controlled trials (RCTs) of non-renal (EXPLORER) and renal (LUNAR) SLE. Areas covered: We review how BCD could be improved to achieve better clinical responses in RA and SLE. Insights into the variability in clinical response to BCD in RA and SLE may help develop new therapeutic strategies. To this end, a literature search was performed using the following terms: rheumatoid arthritis, systemic erythematosus lupus, rituximab and B-cell depletion. Expert commentary: Poor trial design may have, at least partly, contributed to the apparent lack of response to BCD in the two RCTs of patients with SLE. Enhanced B-cell depletion and/or sequential therapy with belimumab may improve clinical response at least in some patients with SLE.
Collapse
Affiliation(s)
- Pedro Mota
- a Department of Internal Medicine , Hospital da Luz , Lisbon , Portugal
| | - Venkat Reddy
- b Centre for Rheumatology, Division of Medicine , University College London , London , UK
| | - David Isenberg
- b Centre for Rheumatology, Division of Medicine , University College London , London , UK
| |
Collapse
|
90
|
Toyoda M, Thomas D, Ahn G, Kahwaji J, Mirocha J, Chu M, Vo A, Suviolahti E, Ge S, Jordan SC. JC polyomavirus viremia and progressive multifocal leukoencephalopathy in human leukocyte antigen-sensitized kidney transplant recipients desensitized with intravenous immunoglobulin and rituximab. Transpl Infect Dis 2016; 17:838-47. [PMID: 26437369 DOI: 10.1111/tid.12465] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 07/27/2015] [Accepted: 09/07/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND Desensitization (DES) with intravenous immunoglobulin (IVIG) + rituximab is effective, safe, and increases the transplantation rate in human leukocyte antigen-sensitized patients. However, reports of progressive multifocal leukoencephalopathy (PML) caused by JC polyomavirus (JCPyV) in autoimmune patients treated with rituximab is concerning. Here, we report on the JCPyV viremia and PML status in kidney transplant patients with/without DES (non-DES). METHODS In total 1195 and 699 DNA samples from plasma in 117 DES (78% lymphocyte-depleting [LyD] induction) and 100 non-DES patients (45% LyD), respectively, were submitted for JCPyV-polymerase chain reaction. Results were compared in both groups. RESULTS No patients in either DES or non-DES developed PML or presented with any neurological symptoms. The JCPyV viremia rate was similar in DES and non-DES patients (3/117 vs. 9/100, P = 0.07). The JCPyV levels were low (median peak levels, 1025 copies/mL) and JCPyV viremia was observed only once during the study period in most patients. All 3 DES patients with JCPyV(+) received 1 dose rituximab and no DES patients with >1 dose rituximab showed JCPyV(+). All 3 JCPyV(+) DES patients received LyD induction, while only 2 of 9 JCPyV(+) non-DES patients did so, and the remaining 7 received non-LyD or no induction. JCPyV in leukocyte was mostly negative in DES and non-DES patients. Immunosuppression in patients with or without JCPyV(+) was similar. BK polyomavirus viremia was observed more commonly in patients with JCPyV(+) than in those without (P < 0.02). CONCLUSIONS Patients with IVIG + rituximab DES followed by transplantation with LyD induction and additional rituximab rarely show JCPyV viremia and appear at low risk for PML.
Collapse
Affiliation(s)
- M Toyoda
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - D Thomas
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - G Ahn
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - J Kahwaji
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - J Mirocha
- Biostatistics Core, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - M Chu
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - A Vo
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - E Suviolahti
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - S Ge
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - S C Jordan
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
91
|
Alping P, Frisell T, Svenningsson A, Piehl F. Reply. Ann Neurol 2016; 80:791-792. [DOI: 10.1002/ana.24767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 08/14/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Peter Alping
- Department of Clinical NeuroscienceKarolinska InstituteStockholm Sweden
| | - Thomas Frisell
- Department of Medicine SolnaKarolinska InstituteStockholm Sweden
| | - Anders Svenningsson
- Department of Clinical SciencesDanderyd Hospital, Karolinska InstituteStockholm Sweden
| | - Fredrik Piehl
- Department of Clinical NeuroscienceKarolinska InstituteStockholm Sweden
| |
Collapse
|
92
|
McGinley MP, Moss BP, Cohen JA. Safety of monoclonal antibodies for the treatment of multiple sclerosis. Expert Opin Drug Saf 2016; 16:89-100. [DOI: 10.1080/14740338.2017.1250881] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Marisa P. McGinley
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brandon P. Moss
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jeffrey A. Cohen
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
93
|
Coclitu C, Constantinescu CS, Tanasescu R. The future of multiple sclerosis treatments. Expert Rev Neurother 2016; 16:1341-1356. [DOI: 10.1080/14737175.2016.1243056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
94
|
Moreno Torres I, García-Merino A. Anti-CD20 monoclonal antibodies in multiple sclerosis. Expert Rev Neurother 2016; 17:359-371. [DOI: 10.1080/14737175.2017.1245616] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Irene Moreno Torres
- Neuroimmunology unit, Neurology department, Puerta de Hierro-Majadahonda University Hospital, Madrid, Spain
| | - Antonio García-Merino
- Neuroimmunology unit, Neurology department, Puerta de Hierro-Majadahonda University Hospital, Madrid, Spain
| |
Collapse
|
95
|
Salzer J, Svenningsson R, Alping P, Novakova L, Björck A, Fink K, Islam-Jakobsson P, Malmeström C, Axelsson M, Vågberg M, Sundström P, Lycke J, Piehl F, Svenningsson A. Rituximab in multiple sclerosis: A retrospective observational study on safety and efficacy. Neurology 2016; 87:2074-2081. [PMID: 27760868 PMCID: PMC5109942 DOI: 10.1212/wnl.0000000000003331] [Citation(s) in RCA: 257] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 06/02/2016] [Indexed: 11/29/2022] Open
Abstract
Objective: To investigate the safety and efficacy of rituximab in multiple sclerosis (MS). Methods: In this retrospective uncontrolled observational multicenter study, off-label rituximab-treated patients with MS were identified through the Swedish MS register. Outcome data were collected from the MS register and medical charts. Adverse events (AEs) grades 2–5 according to the Common Terminology Criteria for Adverse Events were recorded. Results: A total of 822 rituximab-treated patients with MS were identified: 557 relapsing-remitting MS (RRMS), 198 secondary progressive MS (SPMS), and 67 primary progressive MS (PPMS). At baseline, 26.2% had contrast-enhancing lesions (CELs). Patients were treated with 500 or 1,000 mg rituximab IV every 6–12 months, during a mean 21.8 (SD 14.3) months. During treatment, the annualized relapse rates were 0.044 (RRMS), 0.038 (SPMS), and 0.015 (PPMS), and 4.6% of patients displayed CELs. Median Expanded Disability Status Scale remained unchanged in RRMS (p = 0.42) and increased by 0.5 and 1.0 in SPMS and PPMS, respectively (p = 0.10 and 0.25). Infusion-related AEs occurred during 7.8% of infusions and most were mild. A total of 89 AEs grades ≥2 (of which 76 infections) were recorded in 72 patients. No case of progressive multifocal leukoencephalopathy was detected. Conclusions: This is the largest cohort of patients with MS treated with rituximab reported so far. The safety, clinical, and MRI findings in this heterogeneous real-world cohort treated with different doses of rituximab were similar to those reported in previous randomized controlled trials on B-cell depletion therapy in MS. Classification of evidence: This study provides Class IV evidence that for patients with MS, rituximab is safe and effective.
Collapse
Affiliation(s)
- Jonatan Salzer
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden.
| | - Rasmus Svenningsson
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Peter Alping
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Lenka Novakova
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Anna Björck
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Katharina Fink
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Protik Islam-Jakobsson
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Clas Malmeström
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Markus Axelsson
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Mattias Vågberg
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Peter Sundström
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Jan Lycke
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Fredrik Piehl
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Anders Svenningsson
- From the Department of Pharmacology and Clinical Neuroscience (J.S., R.S., P.I.-J., M.V., P.S., A.S.), Umeå University; Departments of Clinical Neuroscience (R.S., P.A., A.B., K.F., F.P.) and Clinical Sciences, Danderyd Hospital (A.S.), Karolinska Institutet, Stockholm; and Department of Clinical Neuroscience (L.N., C.M., M.A., J.L.), Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
96
|
Bittner S, Ruck T, Wiendl H, Grauer OM, Meuth SG. Targeting B cells in relapsing-remitting multiple sclerosis: from pathophysiology to optimal clinical management. Ther Adv Neurol Disord 2016; 10:51-66. [PMID: 28450895 DOI: 10.1177/1756285616666741] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease that is caused by an autoimmune response against central nervous system (CNS) structures. Traditionally considered a T-cell-mediated disorder, the contribution of B cells to the pathogenesis of MS has long been debated. Based on recent promising clinical results from CD20-depleting strategies by three therapeutic monoclonal antibodies in clinical phase II and III trials (rituximab, ocrelizumab and ofatumumab), targeting B cells in MS is currently attracting growing interest among basic researchers and clinicians. Many questions about the role of B and plasma cells in MS remain still unanswered, ranging from the role of specific B-cell subsets and functions to the optimal treatment regimen of B-cell depletion and monitoring thereafter. Here, we will assess our current knowledge of the mechanisms implicating B cells in multiple steps of disease pathology and examine current and future therapeutic approaches for the treatment of MS.
Collapse
Affiliation(s)
- Stefan Bittner
- Department of Neurology, University of Mainz, Mainz, Germany
| | - Tobias Ruck
- Department of Neurology, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany
| | - Oliver M Grauer
- Department of Neurology, University of Münster, Münster, Germany
| | - Sven G Meuth
- Department of Neurology, University of Münster, Münster, Germany
| |
Collapse
|
97
|
Agahozo MC, Peferoen L, Baker D, Amor S. CD20 therapies in multiple sclerosis and experimental autoimmune encephalomyelitis - Targeting T or B cells? Mult Scler Relat Disord 2016; 9:110-7. [PMID: 27645355 DOI: 10.1016/j.msard.2016.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/06/2016] [Accepted: 07/19/2016] [Indexed: 01/03/2023]
Abstract
MS is widely considered to be a T cell-mediated disease although T cell immunotherapy has consistently failed, demonstrating distinct differences with experimental autoimmune encephalomyelitis (EAE), an animal model of MS in which T cell therapies are effective. Accumulating evidence has highlighted that B cells also play key role in MS pathogenesis. The high frequency of oligoclonal antibodies in the CSF, the localization of immunoglobulin in brain lesions and pathogenicity of antibodies originally pointed to the pathogenic role of B cells as autoantibody producing plasma cells. However, emerging evidence reveal that B cells also act as antigen presenting cells, T cell activators and cytokine producers suggesting that the strong efficacy of anti-CD20 antibody therapy observed in people with MS may reduce disease progression by several different mechanisms. Here we review the evidence and mechanisms by which B cells contribute to disease in MS compared to findings in the EAE model.
Collapse
Affiliation(s)
- Marie Colombe Agahozo
- Pathology Department, VU Medical Centre, VU University of Amsterdam, The Netherlands
| | - Laura Peferoen
- Pathology Department, VU Medical Centre, VU University of Amsterdam, The Netherlands
| | - David Baker
- Neuroimmunolgy Unit, Blizard Institute, Barts and the London School of Medicine & Dentistry Queen Mary University of London, United Kingdom
| | - Sandra Amor
- Pathology Department, VU Medical Centre, VU University of Amsterdam, The Netherlands; Neuroimmunolgy Unit, Blizard Institute, Barts and the London School of Medicine & Dentistry Queen Mary University of London, United Kingdom.
| |
Collapse
|
98
|
Affiliation(s)
- Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, 2300 RC Leiden, Netherlands.
| | - Gerdur Gröndal
- Department of Rheumatology, Landspítali, National University Hospital, Center for Rheumatology Research, Reykjavík, Iceland
| |
Collapse
|
99
|
Drug-associated progressive multifocal leukoencephalopathy: a clinical, radiological, and cerebrospinal fluid analysis of 326 cases. J Neurol 2016; 263:2004-21. [PMID: 27401179 PMCID: PMC5037162 DOI: 10.1007/s00415-016-8217-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 02/07/2023]
Abstract
The implementation of a variety of immunosuppressive therapies has made drug-associated progressive multifocal leukoencephalopathy (PML) an increasingly prevalent clinical entity. The purpose of this study was to investigate its diagnostic characteristics and to determine whether differences herein exist between the multiple sclerosis (MS), neoplasm, post-transplantation, and autoimmune disease subgroups. Reports of possible, probable, and definite PML according to the current diagnostic criteria were obtained by a systematic search of PubMed and the Dutch pharmacovigilance database. Demographic, epidemiologic, clinical, radiological, cerebrospinal fluid (CSF), and histopathological features were extracted from each report and differences were compared between the disease categories. In the 326 identified reports, PML onset occurred on average 29.5 months after drug introduction, varying from 14.2 to 37.8 months in the neoplasm and MS subgroups, respectively. The most common overall symptoms were motor weakness (48.6 %), cognitive deficits (43.2 %), dysarthria (26.3 %), and ataxia (24.1 %). The former two also constituted the most prevalent manifestations in each subgroup. Lesions were more often localized supratentorially (87.7 %) than infratentorially (27.4 %), especially in the frontal (64.1 %) and parietal lobes (46.6 %), and revealed enhancement in 27.6 % of cases, particularly in the MS (42.9 %) subgroup. Positive JC virus results in the first CSF sample were obtained in 63.5 %, while conversion after one or more negative outcomes occurred in 13.7 % of cases. 52.2 % of patients died, ranging from 12.0 to 83.3 % in the MS and neoplasm subgroups, respectively. In conclusion, despite the heterogeneous nature of the underlying diseases, motor weakness and cognitive changes were the two most common manifestations of drug-associated PML in all subgroups. The frontal and parietal lobes invariably constituted the predilection sites of drug-related PML lesions.
Collapse
|
100
|
Abstract
Neurologic complications of cancer are common and are frequently life-threatening events. Certain neurologic emergencies occur more frequently in the cancer population, specifically elevated intracranial pressure, epidural cord compression, status epilepticus, ischemic and hemorrhagic stroke, central nervous system infection, and treatment-associated neurologic dysfunction. These emergencies require early diagnosis and prompt treatment to ensure the best possible outcome and are best managed in the intensive care unit. This article reviews the presentation, pathophysiology, and management of the most common causes of acute neurologic decompensation in the patient with cancer.
Collapse
Affiliation(s)
- Andrew L Lin
- 1 Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Edward K Avila
- 1 Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|