101
|
Barth H, Solis M, Lepiller Q, Sueur C, Soulier E, Caillard S, Stoll-Keller F, Fafi-Kremer S. 45 years after the discovery of human polyomaviruses BK and JC: Time to speed up the understanding of associated diseases and treatment approaches. Crit Rev Microbiol 2016; 43:178-195. [DOI: 10.1080/1040841x.2016.1189873] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Heidi Barth
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Morgane Solis
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Quentin Lepiller
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Charlotte Sueur
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Eric Soulier
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Sophie Caillard
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
- Département de Néphrologie et Transplantation, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Françoise Stoll-Keller
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Samira Fafi-Kremer
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| |
Collapse
|
102
|
Bennett CL, Berger JR, Sartor O, Carson KR, Hrushesky WJ, Georgantopoulos P, Raisch DW, Norris LB, Armitage JO. Progressive multi-focal leucoencephalopathy among ibrutinib-treated persons with chronic lymphocytic leukaemia. Br J Haematol 2016; 180:301-304. [PMID: 27649938 DOI: 10.1111/bjh.14322] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Charles L Bennett
- The Southern Network on Adverse Reactions (SONAR) Program, South Carolina College of Pharmacy, Columbia, SC, USA.,The Arnold School of Public Health, University of South Carolina, Columbia, SC, USA.,SC Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Joseph R Berger
- Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Oliver Sartor
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Kenneth R Carson
- Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - William J Hrushesky
- The Southern Network on Adverse Reactions (SONAR) Program, South Carolina College of Pharmacy, Columbia, SC, USA.,The Arnold School of Public Health, University of South Carolina, Columbia, SC, USA.,SC Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.,Oncology Analytics Inc, Plantation, FL, USA
| | - Peter Georgantopoulos
- The Southern Network on Adverse Reactions (SONAR) Program, South Carolina College of Pharmacy, Columbia, SC, USA.,The Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Dennis W Raisch
- College of Pharmacy, Pharmacoeconomics, Epidemiology, Public Policy, Outcomes Research Program, University of New Mexico, Albuquerque, NM, USA
| | - LeAnn B Norris
- The Southern Network on Adverse Reactions (SONAR) Program, South Carolina College of Pharmacy, Columbia, SC, USA
| | | |
Collapse
|
103
|
Li Y, Min W, Li M, Han G, Dai D, Zhang L, Chen X, Wang X, Zhang Y, Yue Z, Liu J. Identification of hub genes and regulatory factors of glioblastoma multiforme subgroups by RNA-seq data analysis. Int J Mol Med 2016; 38:1170-8. [PMID: 27572852 PMCID: PMC5029949 DOI: 10.3892/ijmm.2016.2717] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 08/04/2016] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant brain tumor. This study aimed to identify the hub genes and regulatory factors of GBM subgroups by RNA sequencing (RNA-seq) data analysis, in order to explore the possible mechanisms responsbile for the progression of GBM. The dataset RNASeqV2 was downloaded by TCGA-Assembler, containing 169 GBM and 5 normal samples. Gene expression was calculated by the reads per kilobase per million reads measurement, and nor malized with tag count comparison. Following subgroup classification by the non-negative matrix factorization, the differentially expressed genes (DEGs) were screened in 4 GBM subgroups using the method of significance analysis of microarrays. Functional enrichment analysis was performed by DAVID, and the protein-protein interaction (PPI) network was constructed based on the HPRD database. The subgroup-related microRNAs (miRNAs or miRs), transcription factors (TFs) and small molecule drugs were predicted with predefined criteria. A cohort of 19,515 DEGs between the GBM and control samples was screened, which were predominantly enriched in cell cycle- and immunoreaction-related pathways. In the PPI network, lymphocyte cytosolic protein 2 (LCP2), breast cancer 1 (BRCA1), specificity protein 1 (Sp1) and chromodomain-helicase-DNA-binding protein 3 (CHD3) were the hub nodes in subgroups 1–4, respectively. Paired box 5 (PAX5), adipocyte protein 2 (aP2), E2F transcription factor 1 (E2F1) and cAMP-response element-binding protein-1 (CREB1) were the specific TFs in subgroups 1–4, respectively. miR-147b, miR-770-5p, miR-220a and miR-1247 were the particular miRNAs in subgroups 1–4, respectively. Natalizumab was the predicted small molecule drug in subgroup 2. In conclusion, the molecular regulatory mechanisms of GBM pathogenesis were distinct in the different subgroups. Several crucial genes, TFs, miRNAs and small molecules in the different GBM subgroups were identified, which may be used as potential markers. However, further experimental validations may be required.
Collapse
Affiliation(s)
- Yanan Li
- Department of Neurosurgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Weijie Min
- Department of Neurosurgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Mengmeng Li
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
| | - Guosheng Han
- Department of Neurosurgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Dongwei Dai
- Department of Neurosurgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Lei Zhang
- Department of Neurosurgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Xin Chen
- Department of Neurosurgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Xinglai Wang
- Department of Neurosurgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Yuhui Zhang
- Department of Neurosurgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Zhijian Yue
- Department of Neurosurgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Jianmin Liu
- Department of Neurosurgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
104
|
Melis M, Biagi C, Småbrekke L, Nonino F, Buccellato E, Donati M, Vaccheri A, Motola D. Drug-Induced Progressive Multifocal Leukoencephalopathy: A Comprehensive Analysis of the WHO Adverse Drug Reaction Database. CNS Drugs 2016; 29:879-91. [PMID: 26507833 DOI: 10.1007/s40263-015-0286-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To identify safety signals concerning the association between the use of various drug classes and the onset of progressive multifocal leukoencephalopathy (PML). METHODS All reports containing suspected or interacting PML-related or leukoencephalopathy-related drugs, held in the World Health Organization spontaneous individual case safety reports database as at 1 September 2014, were retrieved. We identified safety signals by analysing the drug-reaction pairs, using the reporting odds ratio as a measure of disproportionality. A safety signal was defined if a drug was reported more than twice in PML cases with a reporting odds ratio >2 and a lower 95 % confidence limit >1. RESULTS We retrieved 2452 reports associated with PML (N = 1612), leukoencephalopathy (N = 835) or both (N = 5), corresponding to 343 different drugs. PML was reported similarly in male and female adults (18-64 years), and almost 30 % of the cases had a fatal outcome. The most frequent Anatomical Therapeutic Chemical (ATC) classification groups concerned antineoplastic agents (23.5 %), antivirals for systemic use (10.1 %) or immunostimulants (4.6 %). Significant disproportionality was found for 88 drugs in the overall analysis (of cases with 'progressive multifocal leukoencephalopathy' or 'leukoencephalopathy' as the Preferred Term), and a new safety signal was identified for 59 active substances (e.g. muromonab-CD3, basiliximab and antithymocyte Ig), as no information on a possible risk of PML was acknowledged in their Summary of Product Characteristics documents. Some safety signals were confirmed also after sensitivity analysis adjustment for several confounding factors (underlying diseases and considering only 'progressive multifocal leukoencephalopathy' as the Preferred Term). CONCLUSION We report a possible association between several drugs and PML that has not been previously described. In addition, we have confirmed previously reported signals in a number of drugs. We highlight the need for follow-up by regulatory agencies.
Collapse
Affiliation(s)
- Mauro Melis
- Unit of Pharmacology, Department of Medical and Surgical Sciences, University of Bologna, via Irnerio 48, 40126, Bologna, Italy
| | - Chiara Biagi
- Unit of Pharmacology, Department of Medical and Surgical Sciences, University of Bologna, via Irnerio 48, 40126, Bologna, Italy
| | - Lars Småbrekke
- Department of Pharmacy, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Francesco Nonino
- Medicines and Medical Devices Area - Health and Social Policies Directorate, Emilia-Romagna Region, via Aldo Moro 21, 40127, Bologna, Italy
| | - Elena Buccellato
- Unit of Pharmacology, Department of Medical and Surgical Sciences, University of Bologna, via Irnerio 48, 40126, Bologna, Italy
| | - Monia Donati
- Unit of Pharmacology, Department of Medical and Surgical Sciences, University of Bologna, via Irnerio 48, 40126, Bologna, Italy
| | - Alberto Vaccheri
- Unit of Pharmacology, Department of Medical and Surgical Sciences, University of Bologna, via Irnerio 48, 40126, Bologna, Italy
| | - Domenico Motola
- Unit of Pharmacology, Department of Medical and Surgical Sciences, University of Bologna, via Irnerio 48, 40126, Bologna, Italy.
| |
Collapse
|
105
|
Lepetit M, Laplaud DA. Effetti collaterali delle bioterapie nella sclerosi multipla e nelle malattie correlate. Neurologia 2016. [DOI: 10.1016/s1634-7072(16)78802-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
106
|
Miskin DP, Herman ST, Ngo LH, Koralnik IJ. Predictors and characteristics of seizures in survivors of progressive multifocal leukoencephalopathy. J Neurovirol 2016; 22:464-71. [PMID: 26676826 PMCID: PMC4937716 DOI: 10.1007/s13365-015-0414-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 11/20/2015] [Accepted: 12/08/2015] [Indexed: 11/29/2022]
Abstract
This study aims to determine the risk factors for epileptogenesis and characteristics of seizures in patients with progressive multifocal leukoencephalopathy (PML) who survive more than 1 year from onset of neurological symptoms (PML survivors). We reviewed clinical data including seizure history and MR imaging studies from PML survivors evaluated at our institution between 1997 and 2014. PML progressors who passed away within 1 year and patients with a history of seizures prior to PML diagnosis were excluded from the analysis. Of 64 PML survivors, 28 (44 %) developed seizures. The median time from the onset of PML symptoms to the first seizure was 5.4 months (range 0-159) and 64 % of patients with seizures had them within the first year. The presence of juxtacortical PML lesions was associated with a relative risk of seizures of 3.5 (p < 0.02; 95 % confidence interval (CI) 1.3-9.4) in multivariate analyses. Of all seizure types, 86 % were focal and 60 % most likely originated from the frontal lobes. Among seizure patients, 89 % required treatment, including one (54 %), two (25 %), or three (10.5 %) antiepileptic drugs. Seizures are a frequent complication in PML and can develop throughout the entire course of the disease. However, late onset seizures did not signify PML relapse. Seizures may require treatment with multiple antiepileptic medications and are a significant co-morbidity in PML.
Collapse
Affiliation(s)
- Dhanashri P. Miskin
- Division of Neuro-Immunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Susan T. Herman
- Division of Epilepsy, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Long H. Ngo
- Division of General Medicine and Primary Care Section for Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Igor J. Koralnik
- Division of Neuro-Immunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| |
Collapse
|
107
|
Cline A, Hill D, Lewallen R, Feldman SR. Current status and future prospects for biologic treatments of psoriasis. Expert Rev Clin Immunol 2016; 12:1273-1287. [DOI: 10.1080/1744666x.2016.1202115] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
108
|
Verma NK, Fazil MHUT, Ong ST, Chalasani MLS, Low JH, Kottaiswamy A, P P, Kizhakeyil A, Kumar S, Panda AK, Freeley M, Smith SM, Boehm BO, Kelleher D. LFA-1/ICAM-1 Ligation in Human T Cells Promotes Th1 Polarization through a GSK3β Signaling-Dependent Notch Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:108-118. [PMID: 27206767 DOI: 10.4049/jimmunol.1501264] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 05/02/2016] [Indexed: 02/09/2025]
Abstract
In this study, we report that the integrin LFA-1 cross-linking with its ligand ICAM-1 in human PBMCs or CD4(+) T cells promotes Th1 polarization by upregulating IFN-γ secretion and T-bet expression. LFA-1 stimulation in PBMCs, CD4(+) T cells, or the T cell line HuT78 activates the Notch pathway by nuclear translocation of cleaved Notch1 intracellular domain (NICD) and upregulation of target molecules Hey1 and Hes1. Blocking LFA-1 by a neutralizing Ab or specific inhibition of Notch1 by a γ-secretase inhibitor substantially inhibits LFA-1/ICAM-1-mediated activation of Notch signaling. We further demonstrate that the Notch pathway activation is dependent on LFA-1/ICAM-1-induced inactivation of glycogen synthase kinase 3β (GSK3β), which is mediated via Akt and ERK. Furthermore, in silico analysis in combination with coimmunoprecipitation assays show an interaction between NICD and GSK3β. Thus, there exists a molecular cross-talk between LFA-1 and Notch1 through the Akt/ERK-GSK3β signaling axis that ultimately enhances T cell differentiation toward Th1. Although clinical use of LFA-1 antagonists is limited by toxicity related to immunosuppression, these findings support the concept that Notch inhibitors could be attractive for prevention or treatment of Th1-related immunologic disorders and have implications at the level of local inflammatory responses.
Collapse
Affiliation(s)
- Navin K Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921; Singapore Eye Research Institute, Singapore 168751;
| | - M H U Turabe Fazil
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| | - Seow Theng Ong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| | | | - Jian Hui Low
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| | | | - Praseetha P
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| | - Atish Kizhakeyil
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| | - Sunil Kumar
- Institute of Life Sciences, Bhubaneshwar, Odisha 751023, India
| | - Aditya K Panda
- Centre for Life Sciences, Central University of Jharkhand, Ranchi 835205, India
| | - Michael Freeley
- Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Sinead M Smith
- Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; and
| | - Bernhard O Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| | - Dermot Kelleher
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921; Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V1Y 1T3, Canada
| |
Collapse
|
109
|
Anderson DJ, Lo DJ, Leopardi F, Song M, Turgeon NA, Strobert EA, Jenkins JB, Wang R, Reimann KA, Larsen CP, Kirk AD. Anti-Leukocyte Function-Associated Antigen 1 Therapy in a Nonhuman Primate Renal Transplant Model of Costimulation Blockade-Resistant Rejection. Am J Transplant 2016; 16:1456-64. [PMID: 26602755 PMCID: PMC5066576 DOI: 10.1111/ajt.13628] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/20/2015] [Accepted: 11/02/2015] [Indexed: 01/25/2023]
Abstract
Costimulation blockade with the fusion protein belatacept provides a desirable side effect profile and improvement in renal function compared with calcineurin inhibition in renal transplantation. This comes at the cost of increased rates of early acute rejection. Blockade of the integrin molecule leukocyte function-associated antigen 1 (LFA-1) has been shown to be an effective adjuvant to costimulation blockade in a rigorous nonhuman primate (NHP) model of islet transplantation; therefore, we sought to test this combination in an NHP renal transplant model. Rhesus macaques received belatacept maintenance therapy with or without the addition of LFA-1 blockade, which was achieved using a murine-derived LFA-1-specific antibody TS1/22. Additional experiments were performed using chimeric rhesus IgG1 (TS1/22R1) or IgG4 (TS1/22R4) variants, each engineered to limit antibody clearance. Despite evidence of proper binding to the target molecule and impaired cellular egress from the intravascular space indicative of a therapeutic effect similar to prior islet studies, LFA-1 blockade failed to significantly prolong graft survival. Furthermore, evidence of impaired protective immunity against cytomegalovirus was observed. These data highlight the difficulties in translating treatment regimens between organ models and suggest that the primarily vascularized renal model is more robust with regard to belatacept-resistant rejection than the islet model.
Collapse
Affiliation(s)
| | - Denise J. Lo
- Emory Transplant Center, Emory University, Atlanta, GA
| | - F. Leopardi
- Department of Surgery, Duke University, Durham, NC
| | | | | | | | | | - Rijian Wang
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | - Keith A. Reimann
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | | | - Allan D. Kirk
- Emory Transplant Center, Emory University, Atlanta, GA
- Department of Surgery, Duke University, Durham, NC
| |
Collapse
|
110
|
Md Yusof MY, Vital EM, Buch MH. B Cell Therapies, Approved and Emerging: a Review of Infectious Risk and Prevention During Use. Curr Rheumatol Rep 2016; 17:65. [PMID: 26290110 DOI: 10.1007/s11926-015-0539-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The development of B cell-targeted biologics represents a major advance in the treatment of autoimmune rheumatic diseases. As with other immunosuppressive agents, risk of infection is a key clinical concern. This review summarises safety data from 15 years of experience of rituximab in autoimmune diseases with a particular focus on opportunistic infection and class-specific complications and infection risk. Rarely, cases of progressive multifocal leucoencephalopathy in rituximab-treated patients (5/100 000) have accumulated over time although no proven causal association has yet been shown. With repeat cycles of therapy, hypogammaglobulinaemia has been observed in a larger proportion of patients and is associated with increased risk of serious infections. The infection profile of the newer B cell-targeted agent, belimumab, in patients with active systemic lupus erythematosus is also discussed. Data from registries are needed to extend insights further and also to evaluate for any impact with the difference in mode of action of belimumab and infection risk in this population.
Collapse
Affiliation(s)
- Md Yuzaiful Md Yusof
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Chapeltown Road, Leeds, LS7 4SA, UK
| | | | | |
Collapse
|
111
|
Sariyer R, De-Simone FI, Gordon J, Sariyer IK. Immune suppression of JC virus gene expression is mediated by SRSF1. J Neurovirol 2016; 22:597-606. [PMID: 26951564 DOI: 10.1007/s13365-016-0432-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 12/14/2022]
Abstract
Progressive multifocal leukoemcephalopathy (PML) is a fatal demyelinating disease caused by the human neurotropic JC virus (JCV). JCV infects the majority of the human population during childhood and establishes a latent/persistent life-long infection. The virus reactivates under immunosuppressive conditions by unknown mechanisms, resulting in productive infection of oligodendrocytes in the central nervous system (CNS). Given the fact that the natural occurrence of PML is strongly associated with immunosuppression, the functional and molecular interaction between glial cells and neuroimmune signaling mediated by soluble immune mediators is likely to play a major role in reactivation of JCV and the progression of the lytic viral life cycle leading to the development of PML. In order to explore the effect of soluble immune mediators secreted by peripheral blood mononuclear cells (PBMCs) on JCV transcription, primary human fetal glial (PHFG) cells were treated with conditioned media from PBMCs. We observed a strong suppression of JCV early as well as late gene transcription in cells treated with conditioned media from induced PBMCs. Using a variety of virological and molecular biological approaches, we demonstrate that immune mediators secreted by PBMCs induce the expression of SRSF1, a strong inhibitor of JCV gene expression, and inhibit the replication of JCV. Our results show that downregulation of SRSF1 in glial cells overcomes the suppression of JCV gene expression and its replication mediated by soluble immune mediators. These findings suggest the presence of a novel immune signaling pathway between glial cells and PBMCs that may control JCV gene expression during the course of viral reactivation.
Collapse
Affiliation(s)
- Rahsan Sariyer
- Department of Neuroscience, Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 North Broad Street, 7th Floor, Philadelphia, PA, 19140, USA
| | - Francesca Isabella De-Simone
- Department of Neuroscience, Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 North Broad Street, 7th Floor, Philadelphia, PA, 19140, USA
| | - Jennifer Gordon
- Department of Neuroscience, Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 North Broad Street, 7th Floor, Philadelphia, PA, 19140, USA
| | - Ilker Kudret Sariyer
- Department of Neuroscience, Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 North Broad Street, 7th Floor, Philadelphia, PA, 19140, USA.
| |
Collapse
|
112
|
Ley K, Rivera-Nieves J, Sandborn WJ, Shattil S. Integrin-based therapeutics: biological basis, clinical use and new drugs. Nat Rev Drug Discov 2016; 15:173-83. [PMID: 26822833 PMCID: PMC4890615 DOI: 10.1038/nrd.2015.10] [Citation(s) in RCA: 307] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Integrins are activatable molecules that are involved in adhesion and signalling. Of the 24 known human integrins, 3 are currently targeted therapeutically by monoclonal antibodies, peptides or small molecules: drugs targeting the platelet αIIbβ3 integrin are used to prevent thrombotic complications after percutaneous coronary interventions, and compounds targeting the lymphocyte α4β1 and α4β7 integrins have indications in multiple sclerosis and inflammatory bowel disease. New antibodies and small molecules targeting β7 integrins (α4β7 and αEβ7 integrins) and their ligands are in clinical development for the treatment of inflammatory bowel diseases. Integrin-based therapeutics have shown clinically significant benefits in many patients, leading to continued medical interest in the further development of novel integrin inhibitors. Of note, almost all integrin antagonists in use or in late-stage clinical trials target either the ligand-binding site or the ligand itself.
Collapse
Affiliation(s)
- Klaus Ley
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle Drive, La Jolla, Califoria 92037, USA, and the Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093 USA
| | - Jesus Rivera-Nieves
- La Jolla Institute for Allergy and the Immunology, 9420 Athena Circle Drive, La Jolla, Califoria 92037, USA, and the Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093 USA
| | - William J Sandborn
- Immunology and the Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093 USA
| | - Sanford Shattil
- Division of Haematology-Oncology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093 USA
| |
Collapse
|
113
|
van Nimwegen JF, Moerman RV, Sillevis Smitt N, Brouwer E, Bootsma H, Vissink A. Safety of treatments for primary Sjögren's syndrome. Expert Opin Drug Saf 2016; 15:513-24. [PMID: 26809028 DOI: 10.1517/14740338.2016.1146676] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Primary Sjögren's syndrome (pSS) is a disabling auto-immune disease, affecting exocrine glands and several organs. AREAS COVERED In this review we analyze the safety of therapies used in pSS. Symptomatic treatment is widely applied due to the good supportive effect and good safety profile. Systemic stimulation of tears and saliva can be successful in pSS. However, cumbersome adverse events can influence the tolerability of this therapy. Evidence for the effectiveness of synthetic DMARDs therapies in pSS is limited, while there is a risk of adverse events. Several studies on biologic DMARD treatment of pSS patients have shown promising efficacy and safety results. EXPERT OPINION The safety of symptomatic treatment of pSS is very good. However, systemic therapy is necessary to achieve long-term relieve and prevention of organ-damage. Synthetic DMARDs have not shown much efficacy in earlier studies, and their benefits do not weigh up to the possible harms, while biologic DMARDs show promising results regarding efficacy and cause mostly mild adverse events. Many questions remain unanswered regarding safety of DMARDs in pSS. There is a need for well designed studies, in which safety should be evaluated in a uniform manner to be able to compare the results between studies.
Collapse
Affiliation(s)
- Jolien F van Nimwegen
- a Department of Rheumatology and Clinical Immunology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Rada V Moerman
- a Department of Rheumatology and Clinical Immunology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Nicole Sillevis Smitt
- b Department of Ophthalmology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Elisabeth Brouwer
- a Department of Rheumatology and Clinical Immunology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Hendrika Bootsma
- a Department of Rheumatology and Clinical Immunology , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Arjan Vissink
- c Department of Oral and Maxillofacial Surgery , University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| |
Collapse
|
114
|
Abstract
Post-transplant lymphoproliferative disorders (PTLDs) are a group of conditions that involve uncontrolled proliferation of lymphoid cells as a consequence of extrinsic immunosuppression after organ or haematopoietic stem cell transplant. PTLDs show some similarities to classic lymphomas in the non-immunosuppressed general population. The oncogenic Epstein-Barr virus (EBV) is a key pathogenic driver in many early-onset cases, through multiple mechanisms. The incidence of PTLD varies with the type of transplant; a clear distinction should therefore be made between the conditions after solid organ transplant and after haematopoietic stem cell transplant. Recipient EBV seronegativity and the intensity of immunosuppression are among key risk factors. Symptoms and signs depend on the localization of the lymphoid masses. Diagnosis requires histopathology, although imaging techniques can provide additional supportive evidence. Pre-emptive intervention based on monitoring EBV levels in blood has emerged as the preferred strategy for PTLD prevention. Treatment of established disease includes reduction of immunosuppression and/or administration of rituximab (a B cell-specific antibody against CD20), chemotherapy and EBV-specific cytotoxic T cells. Despite these strategies, the mortality and morbidity remains considerable. Patient outcome is influenced by the severity of presentation, treatment-related complications and risk of allograft loss. New innovative treatment options hold promise for changing the outlook in the future.
Collapse
|
115
|
Doerfler PA, Nayak S, Corti M, Morel L, Herzog RW, Byrne BJ. Targeted approaches to induce immune tolerance for Pompe disease therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:15053. [PMID: 26858964 PMCID: PMC4729315 DOI: 10.1038/mtm.2015.53] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/04/2015] [Accepted: 11/28/2015] [Indexed: 12/31/2022]
Abstract
Enzyme and gene replacement strategies have developed into viable therapeutic approaches for the treatment of Pompe disease (acid α-glucosidase (GAA) deficiency). Unfortunately, the introduction of GAA and viral vectors encoding the enzyme can lead to detrimental immune responses that attenuate treatment benefits and can impact patient safety. Preclinical and clinical experience in addressing humoral responses toward enzyme and gene therapy for Pompe disease have provided greater understanding of the immunological consequences of the provided therapy. B- and T-cell modulation has been shown to be effective in preventing infusion-associated reactions during enzyme replacement therapy in patients and has shown similar success in the context of gene therapy. Additional techniques to induce humoral tolerance for Pompe disease have been the targeted expression or delivery of GAA to discrete cell types or tissues such as the gut-associated lymphoid tissues, red blood cells, hematopoietic stem cells, and the liver. Research into overcoming preexisting immunity through immunomodulation and gene transfer are becoming increasingly important to achieve long-term efficacy. This review highlights the advances in therapies as well as the improved understanding of the molecular mechanisms involved in the humoral immune response with emphasis on methods employed to overcome responses associated with enzyme and gene therapies for Pompe disease.
Collapse
Affiliation(s)
- Phillip A Doerfler
- Department of Pediatrics, University of Florida , Gainesville, Florida, USA
| | - Sushrusha Nayak
- Department of Medicine, Karolinska Institute , Stockholm, Sweden
| | - Manuela Corti
- Department of Pediatrics, University of Florida , Gainesville, Florida, USA
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida , Gainesville, Florida, USA
| | - Roland W Herzog
- Department of Pediatrics, University of Florida , Gainesville, Florida, USA
| | - Barry J Byrne
- Department of Pediatrics, University of Florida , Gainesville, Florida, USA
| |
Collapse
|
116
|
Alexopoulos H, Biba A, Dalakas MC. Anti-B-Cell Therapies in Autoimmune Neurological Diseases: Rationale and Efficacy Trials. Neurotherapeutics 2016; 13:20-33. [PMID: 26566961 PMCID: PMC4720683 DOI: 10.1007/s13311-015-0402-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
B cells have an ever-increasing role in the etiopathology of a number of autoimmune neurological disorders, acting as antibody-producing cells and, most importantly, as sensors, coordinators, and regulators of the immune response. B cells, among other functions, regulate the T-cell activation process through their participation in antigen presentation and production of cytokines. The availability of monoclonal antibodies or fusion proteins against B-cell surface molecules or B-cell trophic factors bestows a rational approach for treating autoimmune neurological disorders, even when T cells are the main effector cells. This review summarizes basic aspects of B-cell biology, discusses the role(s) of B cells in neurological autoimmunity, and presents anti-B-cell drugs that are either currently on the market or are expected to be available in the near future for treating neurological autoimmune disorders.
Collapse
Affiliation(s)
- Harry Alexopoulos
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Angie Biba
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
117
|
Gafter-Gvili A, Polliack A. Bendamustine associated immune suppression and infections during therapy of hematological malignancies. Leuk Lymphoma 2015; 57:512-9. [PMID: 26696321 DOI: 10.3109/10428194.2015.1110748] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bendamustine is being increasingly used in patients with indolent non-Hodgkin lymphoma, mantle cell lymphoma and chronic lymphocytic leukemia. This review summarizes available evidence regarding the effects of bendamustine on the immune system, examines its role in consequent infections as reported in randomized controlled trials, prospective observational investigations, retrospective studies and individual published case reports. Myelosuppression including lymphopenia occurs relatively frequently after therapy with bendamustine. It is mostly CD4 + T cell counts that are suppressed, yet when given in combination with rituximab, both T cell and B cell depletion have been recorded. In addition, hypogammaglobulinemia after bendamustine therapy has also been reported. Variable infection rates have been documented and these include different bacterial, viral and fungal infections. Finally, we also consider issues relating to the use of prophylactic antibiotics in patients receiving the drug.
Collapse
Affiliation(s)
- Anat Gafter-Gvili
- a Department of Medicine A , Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center , Petah-Tikva , Israel ;,b Sackler School of Medicine , Tel-Aviv , Israel
| | - Aaron Polliack
- c Department of Hematology , Hadassah University Hospital and Hebrew University Medical School , Ein Karem , Jerusalem , Israel
| |
Collapse
|
118
|
Impact of Leukocyte Function-Associated Antigen-1 Blockade on Endogenous Allospecific T Cells to Multiple Minor Histocompatibility Antigen Mismatched Cardiac Allograft. Transplantation 2015; 99:2485-93. [DOI: 10.1097/tp.0000000000000805] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
119
|
Craigie M, Regan P, Otalora YL, Sariyer IK. Molecular interplay between T-Antigen and splicing factor, arginine/serine-rich 1 (SRSF1) controls JC virus gene expression in glial cells. Virol J 2015; 12:196. [PMID: 26596376 PMCID: PMC4657255 DOI: 10.1186/s12985-015-0426-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/15/2015] [Indexed: 01/12/2023] Open
Abstract
Background Human polyomavirus JCV is the etiologic agent of progressive multifocal leukoencephalopathy (PML), a fatal demyelinating disease characterized by lytic infection of glial cells in the central nervous system. PML is seen primarily in immunosuppressed patients and is mainly classified as an AIDS-defining disease. In addition to structural capsid proteins, JCV encodes multiple regulatory proteins, including T-antigen and agnoprotein, which are required for functional lytic infection. Previous studies have suggested that molecular interaction between viral proteins and host factors play an important role in reactivation of JCV and progression of the viral life cycle in glial cells. Recently, serine/arginine rich splicing factor 1 (SRSF1), a cellular alternative splicing factor, was identified as a strong negative regulator of JCV in glial cells. SRSF1 inhibits JCV gene expression and viral replication by directly interacting with viral promoter sequences. Here, we have investigated possible impact of JCV regulatory proteins, T-antigen and agnoprotein, on SRSF1-mediated suppression of JCV gene expression in glial cells. Results Reporter gene analysis has suggested that T-antigen rescues viral transcriptional suppression mediated by SRSF1. Further analyses have revealed that T-antigen promotes viral gene expression by suppressing SRSF1 gene transcription in glial cells. A subsequent ChIP analysis revealed that T-antigen associates with the promoter region of SRSF1 to induce the transcriptional suppression. Conclusions These findings have revealed a molecular interplay between cellular SRSF1 and viral T-antigen in controlling JCV gene expression, and may suggest a novel mechanism of JCV reactivation in patients who are at risk of developing PML.
Collapse
Affiliation(s)
- Michael Craigie
- Department of Neuroscience, Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 North Broad Street, 7th Floor, Philadelphia, PA, 19140, USA.
| | - Patrick Regan
- Department of Neuroscience, Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 North Broad Street, 7th Floor, Philadelphia, PA, 19140, USA.
| | - Yolanda-Lopez Otalora
- Department of Neuroscience, Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 North Broad Street, 7th Floor, Philadelphia, PA, 19140, USA.
| | - Ilker Kudret Sariyer
- Department of Neuroscience, Center for Neurovirology, Temple University Lewis Katz School of Medicine, 3500 North Broad Street, 7th Floor, Philadelphia, PA, 19140, USA.
| |
Collapse
|
120
|
Ravani P, Bonanni A, Rossi R, Caridi G, Ghiggeri GM. Anti-CD20 Antibodies for Idiopathic Nephrotic Syndrome in Children. Clin J Am Soc Nephrol 2015; 11:710-20. [PMID: 26585985 DOI: 10.2215/cjn.08500815] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Rituximab, a chimeric anti-CD20 monoclonal antibody originally licensed for lymphoma, is emerging as a novel steroid-sparing agent for idiopathic nephrotic syndrome in children. The potential use of anti-CD20 monoclonal antibodies in idiopathic nephrotic syndrome has contributed to shifting the view of podocytopathies from T cell-mediated to more complex immunomediated disorders that can benefit from targeting B cells and other mediators of the early immune response. Clinical data on the use of rituximab also have implications on disease management and classification. In this review, we present results of clinical studies that support rituximab as an effective steroid-sparing agent in steroid-dependent idiopathic nephrotic syndrome. Recent randomized controlled trials suggest that potential benefits of rituximab therapy in steroid-dependent forms of idiopathic nephrotic syndrome vary depending on whether children are dependent on steroids alone or on both steroids and calcineurin inhibitors, with greater probabilities to achieve drug-free remission in the former group. Multiple-drug dependence may identify a different disease state with different prognosis and treatment options. Insufficient data are available on optimal use of rituximab as a maintenance steroid-sparing agent in these steroid-sensitive forms of the disease, including how often and for how long rituximab infusions should be repeated to maximize expected benefits and minimize potential harms. Finally, one randomized controlled trial in children with steroid-resistant idiopathic nephrotic syndrome yielded negative results. New anti-CD20 antibodies are under study in this patient population.
Collapse
Affiliation(s)
- Pietro Ravani
- Division of Nephrology, University of Calgary, Calgary, Alberta, Canada; and
| | - Alice Bonanni
- Division of Nephrology, Dialysis, Transplantation and
| | - Roberta Rossi
- Division of Nephrology, Dialysis, Transplantation and
| | | | - Gian Marco Ghiggeri
- Division of Nephrology, Dialysis, Transplantation and Laboratory on Pathophysiology of Uremia, Giannina Gaslini Children's Hospital, Genoa, Italy
| |
Collapse
|
121
|
Abstract
Neuromyelitis optica (NMO) is an autoimmune demyelinating condition of the central nervous system often associated with aquaporin-4 (AQP4) autoantibodies manifesting as severe optic neuritis and long segment myelitis with tendency to relapse. Seronegative patients and who do not meet the NMO criteria are classified as having NMO Spectrum Disorder (NMOSD), but are treated identically to clinically definite NMO. Acute relapse is treated with intravenous methylprednisolone for 5 days with or without subsequent treatment with plasma exchange (PE). This must be followed by oral steroid to prevent rebound worsening and further relapse. For relapse prevention, immunosuppressive agents that have been found to be effective are azathioprine, rituximab, mycophenolate mofetil, methotrexate, and mitoxantrone; although none of which have been validated in randomized, controlled trial. Some patients do relapse with monotherapy, and switching to more effective agent or use of combination therapy is beneficial in such situation. There is no consensus about the duration of preventive therapy, but generally 2-3 years of relapse-free period is considered the minimum, taking into account the risks of long-term toxicity of these agents.
Collapse
Affiliation(s)
- Atanu Biswas
- Department of Neurology, Bangur Institute of Neurosciences, Kolkata, India
| | - Arabinda Mukherjee
- Department of Neurology, Vivekananda Institute of Medical Sciences, Kolkata, West Bengal, India
| |
Collapse
|
122
|
Jelcic I, Combaluzier B, Jelcic I, Faigle W, Senn L, Reinhart BJ, Ströh L, Nitsch RM, Stehle T, Sospedra M, Grimm J, Martin R. Broadly neutralizing human monoclonal JC polyomavirus VP1-specific antibodies as candidate therapeutics for progressive multifocal leukoencephalopathy. Sci Transl Med 2015; 7:306ra150. [PMID: 26400911 DOI: 10.1126/scitranslmed.aac8691] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/27/2015] [Indexed: 12/15/2022]
Abstract
In immunocompromised individuals, JC polyomavirus (JCPyV) may mutate and gain access to the central nervous system resulting in progressive multifocal leukoencephalopathy (PML), an often fatal opportunistic infection for which no treatments are currently available. Despite recent progress, the contribution of JCPyV-specific humoral immunity to controlling asymptomatic infection throughout life and to eliminating JCPyV from the brain is poorly understood. We examined antibody responses against JCPyV major capsid protein VP1 (viral protein 1) variants in the serum and cerebrospinal fluid (CSF) of healthy donors (HDs), JCPyV-positive multiple sclerosis patients treated with the anti-VLA-4 monoclonal antibody natalizumab (NAT), and patients with NAT-associated PML. Before and during PML, CSF antibody responses against JCPyV VP1 variants show "recognition holes"; however, upon immune reconstitution, CSF antibody titers rise, then recognize PML-associated JCPyV VP1 variants, and may be involved in elimination of the virus. We therefore reasoned that the memory B cell repertoire of individuals who recovered from PML could be a source for the molecular cloning of broadly neutralizing antibodies for passive immunization. We generated a series of memory B cell-derived JCPyV VP1-specific human monoclonal antibodies from HDs and a patient with NAT-associated PML-immune reconstitution inflammatory syndrome (IRIS). These antibodies exhibited diverse binding affinity, cross-reactivity with the closely related BK polyomavirus, recognition of PML-causing VP1 variants, and JCPyV neutralization. Almost all antibodies with exquisite specificity for JCPyV, neutralizing activity, recognition of all tested JCPyV PML variants, and high affinity were derived from one patient who had recovered from PML. These antibodies are promising drug candidates for the development of a treatment of PML.
Collapse
Affiliation(s)
- Ivan Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | | | - Ilijas Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Wolfgang Faigle
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Luzia Senn
- Neurimmune Holding AG, 8952 Schlieren, Switzerland
| | - Brenda J Reinhart
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Luisa Ströh
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany
| | - Roger M Nitsch
- Neurimmune Holding AG, 8952 Schlieren, Switzerland. Division of Psychiatry Research, University of Zurich, 8952 Schlieren, Switzerland
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany. Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Mireia Sospedra
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Jan Grimm
- Neurimmune Holding AG, 8952 Schlieren, Switzerland.
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland.
| |
Collapse
|
123
|
Doubek M, Šmída M. Treatment of chronic lymphocytic leukemia with monoclonal antibodies, where are we heading? Expert Rev Hematol 2015; 8:743-64. [PMID: 26306923 DOI: 10.1586/17474086.2015.1079123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is the most prevalent leukemia in the western world and monoclonal antibodies (mAbs) are important part of CLL treatment. The goal of this article was to summarize current literature on the position of mAbs in CLL treatment and to mention factors influencing effectiveness of mAbs in CLL. Several new mAbs have been developed and investigated in CLL over the past few years. Mainly anti-CD20 monoclonal antibodies are still used routinely in CLL therapy. Unfortunately, the clinical application of mAbs needs to be further improved. Novel combinations and sequences of mAbs with other compounds need to be studied in clinical trials in order to increase overall response rate and prolong remission duration. Mechanisms of action of mAbs or mechanisms of resistance to mAbs have to be also investigated to predict effectiveness of mAb in particular patient.
Collapse
Affiliation(s)
- Michael Doubek
- a 1 Department of Internal Medicine - Hematology and Oncology, University Hospital and Faculty of Medicine, Brno, Czech Republic.,b 2 Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Michal Šmída
- b 2 Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| |
Collapse
|
124
|
D'Ambrosio A, Pontecorvo S, Colasanti T, Zamboni S, Francia A, Margutti P. Peripheral blood biomarkers in multiple sclerosis. Autoimmun Rev 2015; 14:1097-110. [PMID: 26226413 DOI: 10.1016/j.autrev.2015.07.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/23/2015] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis is the most common autoimmune disorder affecting the central nervous system. The heterogeneity of pathophysiological processes in MS contributes to the highly variable course of the disease and unpredictable response to therapies. The major focus of the research on MS is the identification of biomarkers in biological fluids, such as cerebrospinal fluid or blood, to guide patient management reliably. Because of the difficulties in obtaining spinal fluid samples and the necessity for lumbar puncture to make a diagnosis has reduced, the research of blood-based biomarkers may provide increasingly important tools for clinical practice. However, currently there are no clearly established MS blood-based biomarkers. The availability of reliable biomarkers could radically alter the management of MS at critical phases of the disease spectrum, allowing for intervention strategies that may prevent evolution to long-term neurological disability. This article provides an overview of this research field and focuses on recent advances in blood-based biomarker research.
Collapse
Affiliation(s)
- Antonella D'Ambrosio
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Pontecorvo
- Multiple Sclerosis Center of Department of Neurology and Psychiatry of "Sapienza" University of Rome, Italy
| | - Tania Colasanti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Silvia Zamboni
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Ada Francia
- Multiple Sclerosis Center of Department of Neurology and Psychiatry of "Sapienza" University of Rome, Italy
| | - Paola Margutti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
125
|
Tan HK, Tang CH. A hitherto unrecognised case of progressive multifocal leukoencephalopathy. BMJ Case Rep 2015. [PMID: 26206778 DOI: 10.1136/bcr-2014-208255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
A 65-year-old man presented with a 6-week history of increasing confusion with associated cognitive decline. Empirical antibiotic cover for a urinary tract infection was prescribed with no response. After exhaustive radiological and pathological investigations were carried out, a diagnosis of John Cunningham virus encephalopathy was made. This case report describes the varied clinical presentation of symptoms and their management options, along with the latest clinical opinions behind them.
Collapse
Affiliation(s)
- Huey Kuan Tan
- Department of Medicine, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Chun Hong Tang
- Department of Trauma and Orthopaedics, Nottingham University Hospitals NHS Trust, Nottingham, UK
| |
Collapse
|
126
|
Antiviral prophylaxis in patients with solid tumours and haematological malignancies--update of the Guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society for Hematology and Medical Oncology (DGHO). Ann Hematol 2015; 94:1441-50. [PMID: 26193852 PMCID: PMC4525190 DOI: 10.1007/s00277-015-2447-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/06/2015] [Indexed: 01/17/2023]
Abstract
Reactivation of viral infections is common in patients with solid tumour or haematological malignancy. Incidence and severity depend on the extent of cellular immunosuppression. Antiviral prophylaxis may be effective to prevent viral reactivation. In 2006, the Infectious Diseases Working Party of German Society for Hematology and Medical Oncology (DGHO) published guidelines for antiviral prophylaxis in these patient populations. Here, we present an update of these guidelines for patients with solid and haematological malignancies undergoing antineoplastic treatment but not allogeneic stem cell transplantation. Relevant literature for reactivation of different viruses (herpes simplex virus (HSV), varicella zoster virus (VZV), hepatitis B virus (HBV) and respiratory viruses) is discussed to provide evidence-based recommendations for clinicians taking care of this patient population. We recommend a risk-adapted approach with (val)acyclovir against HSV and VZV in patients treated with alemtuzumab, bortezomib or purine analogues. Seasonal vaccination against influenza is recommended for all patients with solid or haematological malignancies regardless of antineoplastic therapy. Hepatitis B screening is recommended in lymphoproliferative disorders, acute leukaemia, and breast cancer, and during treatment with monoclonal anti-B-cell antibodies, anthracyclines, steroids and in autologous stem cell transplantation. In those with a history of hepatitis B prophylactic lamivudine, entecavir or nucleotide analogues as adefovir are recommended to prevent reactivation.
Collapse
|
127
|
Gupta M, Jafri K, Sharim R, Silverman S, Sindher SB, Shahane A, Kwan M. Immune reconstitution inflammatory syndrome associated with biologic therapy. Curr Allergy Asthma Rep 2015; 15:499. [PMID: 25504263 DOI: 10.1007/s11882-014-0499-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The use of biologics in the treatment of autoimmune disease, cancer, and other immune conditions has revolutionized medical care in these areas. However, there are drawbacks to the use of these medications including increased susceptibility to opportunistic infections. One unforeseen risk once opportunistic infection has occurred with biologic use is the onset of immune reconstitution inflammatory syndrome (IRIS) upon drug withdrawal. Although originally described in human immunodeficiency virus (HIV) patients receiving highly active antiretroviral therapy, it has become clear that IRIS may occur when recovery of immune function follows opportunistic infection in the setting of previous immune compromise/suppression. In this review, we draw attention to this potential pitfall on the use of biologic drugs.
Collapse
Affiliation(s)
- Malika Gupta
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
128
|
Use of Alefacept for Preconditioning in Multiply Transfused Pediatric Patients with Nonmalignant Diseases. Biol Blood Marrow Transplant 2015; 21:1845-52. [PMID: 26095669 DOI: 10.1016/j.bbmt.2015.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/04/2015] [Indexed: 12/20/2022]
Abstract
Transfusion-related alloimmunization is a potent barrier to the engraftment of allogeneic hematopoietic stem cells in patients with nonmalignant diseases (NMDs). Memory T cells, which drive alloimmunization, are relatively resistant to commonly used conditioning agents. Alefacept, a recombinant leukocyte function antigen-3/IgG1 fusion protein, targets CD2 and selectively depletes memory versus naive T cells. Three multiply transfused pediatric patients with NMD received a short course of high-dose i.v. alefacept (.25 mg/kg/dose on days -40 and -9 and .5 mg/kg/dose on days -33, -26, -19, and -12) before undergoing unrelated allogeneic transplant in the setting of reduced-intensity pretransplant conditioning and calcineurin inhibitor-based post-transplant graft-versus-host disease (GVHD) prophylaxis. Alefacept infusions were well tolerated in all patients. Peripheral blood flow cytometry was performed at baseline and during and after alefacept treatment. As expected, after the 5 weekly alefacept doses, each patient demonstrated selective loss of CD2(hi)/CCR7(-)/CD45RA(-) effector memory (Tem) and CD2(hi)/CCR7(+)/CD45RA(-) central memory (Tcm) CD4(+) and CD8(+) T cells with relative preservation of the CD2(lo) Tem and Tcm subpopulations. In addition, depletion of CD2(+) natural killer (NK) cells also occurred. Neutrophil recovery was rapid, and all 3 patients had 100% sorted (CD3/CD33) peripheral blood donor chimerism by day +100. Immune reconstitution (by absolute neutrophil, monocyte, and lymphocyte counts) was comparable with a cohort of historical control patients. All 3 patients developed GVHD but are all now off immune suppression and >2 years post-transplant with stable full-donor engraftment. These results suggest that alefacept at higher dosing can deplete both memory T cells and NK cells and that incorporating CD2-targeted depletion into a reduced-intensity transplant regimen is feasible and safe in heavily transfused patients.
Collapse
|
129
|
IFN-Gamma Inhibits JC Virus Replication in Glial Cells by Suppressing T-Antigen Expression. PLoS One 2015; 10:e0129694. [PMID: 26061652 PMCID: PMC4465661 DOI: 10.1371/journal.pone.0129694] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/12/2015] [Indexed: 12/04/2022] Open
Abstract
Objective Patients undergoing immune modulatory therapies for the treatment of autoimmune diseases such as multiple sclerosis, and individuals with an impaired-immune system, most notably AIDS patients, are in the high risk group of developing progressive multifocal leukoencephalopathy (PML), an often lethal disease of the brain characterized by lytic infection of oligodendrocytes in the central nervous system (CNS) with JC virus (JCV). The immune system plays an important regulatory role in controlling JCV reactivation from latent sites by limiting viral gene expression and replication. However, little is known regarding the molecular mechanisms responsible for this regulation. Methods and Results Here, we investigated the impact of soluble immune mediators secreted by activated PBMCs on viral replication and gene expression by cell culture models and molecular virology techniques. Our data revealed that viral gene expression and viral replication were suppressed by soluble immune mediators. Further studies demonstrated that soluble immune mediators secreted by activated PBMCs inhibit viral replication induced by T-antigen, the major viral regulatory protein, by suppressing its expression in glial cells. This unexpected suppression of T-antigen was mainly associated with the suppression of translational initiation. Cytokine/chemokine array studies using conditioned media from activated PBMCs revealed several candidate cytokines with possible roles in this regulation. Among them, only IFN-γ showed a robust inhibition of T-antigen expression. While potential roles for IFN-β, and to a lesser extent IFN-α have been described for JCV, IFN-γ has not been previously implicated. Further analysis of IFN-γ signaling pathway revealed a novel role of Jak1 signaling in control of viral T-antigen expression. Furthermore, IFN-γ suppressed JCV replication and viral propagation in primary human fetal glial cells, and showed a strong anti-JCV activity. Conclusions Our results suggest a novel role for IFN-γ in the regulation of JCV gene expression via downregulation of the major viral regulatory protein, T-antigen, and provide a new avenue of research to understand molecular mechanisms for downregulation of viral reactivation that may lead to development of novel strategies for the treatment of PML.
Collapse
|
130
|
Haley SA, O'Hara BA, Nelson CDS, Brittingham FLP, Henriksen KJ, Stopa EG, Atwood WJ. Human polyomavirus receptor distribution in brain parenchyma contrasts with receptor distribution in kidney and choroid plexus. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2246-58. [PMID: 26056932 DOI: 10.1016/j.ajpath.2015.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/21/2015] [Accepted: 04/14/2015] [Indexed: 11/25/2022]
Abstract
The human polyomavirus, JCPyV, is the causative agent of progressive multifocal leukoencephalopathy, a rare demyelinating disease that occurs in the setting of prolonged immunosuppression. After initial asymptomatic infection, the virus establishes lifelong persistence in the kidney and possibly other extraneural sites. In rare instances, the virus traffics to the central nervous system, where oligodendrocytes, astrocytes, and glial precursors are susceptible to lytic infection, resulting in progressive multifocal leukoencephalopathy. The mechanisms by which the virus traffics to the central nervous system from peripheral sites remain unknown. Lactoseries tetrasaccharide c (LSTc), a pentasaccharide containing a terminal α2,6-linked sialic acid, is the major attachment receptor for polyomavirus. In addition to LSTc, type 2 serotonin receptors are required for facilitating virus entry into susceptible cells. We studied the distribution of virus receptors in kidney and brain using lectins, antibodies, and labeled virus. The distribution of LSTc, serotonin receptors, and virus binding sites overlapped in kidney and in the choroid plexus. In brain parenchyma, serotonin receptors were expressed on oligodendrocytes and astrocytes, but these cells were negative for LSTc and did not bind virus. LSTc was instead found on microglia and vascular endothelium, to which virus bound abundantly. Receptor distribution was not changed in the brains of patients with progressive multifocal leukoencephalopathy. Virus infection of oligodendrocytes and astrocytes during disease progression is LSTc independent.
Collapse
Affiliation(s)
- Sheila A Haley
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Bethany A O'Hara
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Christian D S Nelson
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Frances L P Brittingham
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Kammi J Henriksen
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Edward G Stopa
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Walter J Atwood
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island.
| |
Collapse
|
131
|
Durali D, de Goër de Herve MG, Gasnault J, Taoufik Y. B cells and progressive multifocal leukoencephalopathy: search for the missing link. Front Immunol 2015; 6:241. [PMID: 26042124 PMCID: PMC4437032 DOI: 10.3389/fimmu.2015.00241] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/05/2015] [Indexed: 12/23/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a deadly demyelinating disease due to JC virus (JCV) replication in the brain. PML classically occurs in patients with severe immunodepression, and cases have recently been linked to therapeutic monoclonal antibodies such as natalizumab and also rituximab, which depletes B cells. B cells appear to play a complex role in the pathogenesis of PML. They may act as a viral reservoir and as a vector for viral dissemination in the central nervous system. Anti-JCV antibody responses appear to have a limited effect on JCV replication in the brain. However, accumulating evidence suggests that B cells may considerably influence T cell responses through their cytokine secretion. This immunomodulatory function of B cells may play an important role in the control of JCV infection and in the pathogenesis of PML, including rituximab-induced PML.
Collapse
Affiliation(s)
- Deniz Durali
- Immunology Research Laboratory, Department of Medical Microbiology, School of Medicine, Istanbul Medipol University , Istanbul , Turkey
| | | | - Jacques Gasnault
- IMVA-INSERM U1184, Department of Immunology, Bicetre Hospital, University Paris-sud , Le Kremlin-Bicêtre , France
| | - Yassine Taoufik
- IMVA-INSERM U1184, Department of Immunology, Bicetre Hospital, University Paris-sud , Le Kremlin-Bicêtre , France
| |
Collapse
|
132
|
Zanoni BC, Gandhi RT. Update on opportunistic infections in the era of effective antiretroviral therapy. Infect Dis Clin North Am 2015; 28:501-18. [PMID: 25151568 DOI: 10.1016/j.idc.2014.05.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite enormous improvements in effectiveness of treatment for HIV infection, opportunistic infections continue to occur in those who have not yet been diagnosed with HIV and in those who are not receiving antiretroviral therapy. This review focuses on tuberculosis and cryptococcal infections, the most common opportunistic infections (OIs) in patients living with human immunodeficiency virus infection around the world, as well as on new developments in progressive multifocal leukoencephalopathy and pneumocystis pneumonia. In the sections on these conditions, updates on diagnosis, treatment, and complications, as well as information on when to start antiretroviral therapy is provided. The article concludes with a discussion of new data on 2 vaccine-preventable OIs, human papillomavirus and varicella-zoster virus.
Collapse
Affiliation(s)
- Brian C Zanoni
- Infectious Diseases Division, Massachusetts General Hospital, GRJ 504, 55 Fruit Street, Boston, MA 02114, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Rajesh T Gandhi
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Infectious Diseases Division and Ragon Institute, Massachusetts General Hospital, GRJ 504, 55 Fruit Street, Boston, MA 02114, USA.
| |
Collapse
|
133
|
The Greater Affinity of JC Polyomavirus Capsid for α2,6-Linked Lactoseries Tetrasaccharide c than for Other Sialylated Glycans Is a Major Determinant of Infectivity. J Virol 2015; 89:6364-75. [PMID: 25855729 DOI: 10.1128/jvi.00489-15] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 03/30/2015] [Indexed: 01/04/2023] Open
Abstract
UNLABELLED The human JC polyomavirus (JCPyV) establishes an asymptomatic, persistent infection in the kidneys of the majority of the population and is the causative agent of the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML) in immunosuppressed individuals. The Mad-1 strain of JCPyV, a brain isolate, was shown earlier to require α2,6-linked sialic acid on the lactoseries tetrasaccharide c (LSTc) glycan for attachment to host cells. In contrast, a JCPyV kidney isolate type 3 strain, WT3, has been reported to interact with sialic acid-containing gangliosides, but the role of these glycans in JCPyV infection has remained unclear. To help rationalize these findings and probe the effects of strain-specific differences on receptor binding, we performed a comprehensive analysis of the glycan receptor specificities of these two representative JCPyV strains using high-resolution X-ray crystallography and nuclear magnetic resonance (NMR) spectroscopy, and correlated these data with the results of infectivity assays. We show here that capsid proteins of Mad-1 and WT3 JCPyV can both engage LSTc as well as multiple sialylated gangliosides. However, the binding affinities exhibit subtle differences, with the highest affinity observed for LSTc. Engagement of LSTc is a prerequisite for functional receptor engagement, while the more weakly binding gangliosides are not required for productive infection. Our findings highlight the complexity of virus-carbohydrate interactions and demonstrate that subtle differences in binding affinities, rather than the binding event alone, help determine tissue tropism and viral pathogenesis. IMPORTANCE Viral infection is initiated by attachment to receptors on host cells, and this event plays an important role in viral disease. We investigated the receptor-binding properties of human JC polyomavirus (JCPyV), a virus that resides in the kidneys of the majority of the population and can cause the fatal demyelinating disease progressive multifocal leukoencephalopathy (PML) in the brains of immunosuppressed individuals. JCPyV has been reported to interact with multiple carbohydrate receptors, and we sought to clarify how the interactions between JCPyV and cellular carbohydrate receptors influenced infection. Here we demonstrate that JCPyV can engage numerous sialylated carbohydrate receptors. However, the virus displays preferential binding to LSTc, and only LSTc mediates a productive infection. Our findings demonstrate that subtle differences in binding affinity, rather than receptor engagement alone, are a key determinant of viral infection.
Collapse
|
134
|
Louthrenoo W. Treatment considerations in patients with concomitant viral infection and autoimmune rheumatic diseases. Best Pract Res Clin Rheumatol 2015; 29:319-42. [DOI: 10.1016/j.berh.2015.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 05/08/2015] [Indexed: 12/17/2022]
|
135
|
Doerfler PA, Nayak S, Herzog RW, Morel L, Byrne BJ. BAFF blockade prevents anti-drug antibody formation in a mouse model of Pompe disease. Clin Immunol 2015; 158:140-7. [PMID: 25842186 DOI: 10.1016/j.clim.2015.03.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 03/24/2015] [Accepted: 03/25/2015] [Indexed: 01/09/2023]
Abstract
Antibodies formed against the therapeutic protein are a life-threatening complication that arises during enzyme replacement therapy for Pompe disease (acid α-glucosidase deficiency; GAA). To provide an effective alternative to current practices, we investigated the capacity of anti-B-cell activating factor (BAFF) as a novel drug candidate to prevent antibody formation in a Pompe disease mouse model. A BAFF-neutralizing antibody was administered prophylactically and with maintenance doses in association with enzyme replacement therapy using recombinant human GAA in Gaa(-/-) mice. BAFF blockade delayed antibody production and increased GAA activity within tissues with protection from anaphylaxis. Anti-BAFF also resolved antibody formation during an immune response and precluded the maturation of antibody secreting cells from entering the bone marrow compartment. This treatment modality may therefore be a viable alternative for the clinical management of antibody formation for Pompe disease and has potential use against antibody formation in other protein replacement therapies.
Collapse
Affiliation(s)
| | - Sushrusha Nayak
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Roland W Herzog
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Laurence Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Barry J Byrne
- Department of Pediatrics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
136
|
Parida JR, Misra DP, Wakhlu A, Agarwal V. Is non-biological treatment of rheumatoid arthritis as good as biologics? World J Orthop 2015; 6:278-283. [PMID: 25793168 PMCID: PMC4363810 DOI: 10.5312/wjo.v6.i2.278] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/10/2014] [Accepted: 09/06/2014] [Indexed: 02/06/2023] Open
Abstract
The management of rheumatoid arthritis (RA) in the past three decades has undergone a paradigm shift from symptomatic relief to a "treat-to-target" approach. This has been possible through use of various conventional and biologic disease modifying anti-rheumatic drugs (DMARDs) which target disease pathogenesis at a molecular level. Cost and infection risk preclude regular use of biologics in resource-constrained settings. In the recent years, evidence has emerged that combination therapy with conventional DMARDs is not inferior to biologics in the management of RA and is a feasible cost-effective option.
Collapse
|
137
|
Dhalla F, Murray S, Sadler R, Chaigne-Delalande B, Sadaoka T, Soilleux E, Uzel G, Miller J, Collins GP, Hatton CSR, Bhole M, Ferry B, Chapel HM, Cohen JI, Patel SY. Identification of a novel mutation in MAGT1 and progressive multifocal leucoencephalopathy in a 58-year-old man with XMEN disease. J Clin Immunol 2015; 35:112-8. [PMID: 25504528 PMCID: PMC6328310 DOI: 10.1007/s10875-014-0116-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 11/25/2014] [Indexed: 10/24/2022]
Abstract
XMEN disease (X-linked immunodeficiency with Magnesium defect, Epstein-Barr virus infection and Neoplasia) is a novel primary immune deficiency caused by mutations in MAGT1 and characterised by chronic infection with Epstein-Barr virus (EBV), EBV-driven lymphoma, CD4 T-cell lymphopenia, and dysgammaglobulinemia [1]. Functional studies have demonstrated roles for magnesium as a second messenger in T-cell receptor signalling [1], and for NKG2D expression and consequently NK- and CD8 T-cell cytotoxicity [2]. 7 patients have been described in the literature; the oldest died at 45 years and was diagnosed posthumously [1-3]. We present the case of a 58-year-old Caucasian gentleman with a novel mutation in MAGT1 with the aim of adding to the phenotype of this newly described disease by detailing his clinical course over more than 20 years.
Collapse
Affiliation(s)
- Fatima Dhalla
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Late Occurrence of PML in a Patient Treated for Lymphoma with Immunomodulatory Chemotherapies, Bendamustine, Rituximab, and Ibritumomab Tiuxetan. Case Rep Neurol Med 2015; 2015:892047. [PMID: 25705531 PMCID: PMC4326342 DOI: 10.1155/2015/892047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 01/07/2015] [Accepted: 01/15/2015] [Indexed: 11/18/2022] Open
Abstract
PML caused by John Cunningham (JC) virus is a rare but an increasingly recognized entity. With the advent of newer immunomodulatory therapies with monoclonal antibodies, there is an increasing incidence of PML. Initially concern was restricted to patients treated for multiple sclerosis with natalizumab but more case reports are being reported during treatment for other conditions like Crohn's disease and lymphoma with agents such as rituximab. We report the case of a 66-year-old woman who developed PML a year after completion of therapy with rituximab, ibritumomab, and bendamustine.
Collapse
|
139
|
Abstract
A 55-year-old woman with known relapsing-remitting multiple sclerosis (RRMS) on natalizumab (Tysabri®) for 3 years was admitted to the hospital with worsening word-finding difficulties and gait instability. Neurologic examination revealed right hemianopia, right arm hemiplegia, right-sided sensory loss, and global aphasia. The patient underwent MRI and PET imaging with concurrent electroencephalogram. She was subsequently diagnosed with natalizumab-induced progressive multifocal leukoencephalopathy (PML) and treated with plasmapheresis, intravenous immunoglobulin, and high-dose intravenous steroids. Steroids were continued over a 3-month hospital course and tapered upon discharge. Speech, arm strength, and ambulation have since improved.
Collapse
|
140
|
Lenert A, Lenert P. Current and emerging treatment options for ANCA-associated vasculitis: potential role of belimumab and other BAFF/APRIL targeting agents. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:333-47. [PMID: 25609919 PMCID: PMC4294650 DOI: 10.2147/dddt.s67264] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) comprises several clinical entities with diverse clinical presentations, outcomes, and nonunifying pathogenesis. AAV has a clear potential for relapses, and shows unpredictable response to treatment. Cyclophosphamide-based therapies have remained the hallmark of induction therapy protocols for more than four decades. Recently, B-cell depleting therapy with the anti-CD20 antibody rituximab has proved beneficial in AAV, leading to Food and Drug Administration approval of rituximab in combination with corticosteroids for the treatment of AAV in adults. Rituximab for ANCA-associated vasculitis and other clinical trials provided clear evidence that rituximab was not inferior to cyclophosphamide for remission induction, and rituximab appeared even more beneficial in patients with relapsing disease. This raised hopes that other B-cell-targeted therapies directed either against CD19, CD20, CD22, or B-cell survival factors, B-cell activating factor of the tumor necrosis factor family (BAFF) and a proliferation-inducing ligand could also be beneficial for the management of AAV. BAFF neutralization with the fully humanized monoclonal antibody belimumab has already shown success in human systemic lupus erythematosus and, along with another anti-BAFF reagent blisibimod, is currently undergoing Phase II and III clinical trials in AAV. Local production of BAFF in granulomatous lesions and elevated levels of serum BAFF in AAV provide a rationale for BAFF-targeted therapies not only in AAV but also in other forms of vasculitis such as Behcet’s disease, large-vessel vasculitis, or cryoglobulinemic vasculitis secondary to chronic hepatitis C infection. BAFF-targeted therapies have a very solid safety profile, and may have an additional benefit of preferentially targeting newly arising autoreactive B cells over non-self-reactive B cells.
Collapse
Affiliation(s)
- Aleksander Lenert
- Division of Rheumatology, University of Kentucky, Kentucky Clinic, Lexington, KY, USA
| | - Petar Lenert
- Division of Immunology, Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
141
|
Frost EL, Lukacher AE. The importance of mouse models to define immunovirologic determinants of progressive multifocal leukoencephalopathy. Front Immunol 2015; 5:646. [PMID: 25601860 PMCID: PMC4283601 DOI: 10.3389/fimmu.2014.00646] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 12/03/2014] [Indexed: 12/02/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a severely debilitating and often fatal demyelinating disease of the central nervous system (CNS) in immunosuppressed individuals caused by JC polyomavirus (JCV), a ubiquitous human pathogen. Demyelination results from lytically infected oligodendrocytes, whose clearance is impaired in the setting of depressed JCV-specific T cell-mediated CNS surveillance. Although mutations in the viral capsid and genomic rearrangements in the viral non-coding region appear to set the stage for PML in the immunosuppressed population, mechanisms of demyelination and CNS antiviral immunity are poorly understood in large part due to absence of a tractable animal model that mimics PML neuropathology in humans. Early studies using mouse polyomavirus (MPyV) in T cell-deficient mice demonstrated productive viral replication in the CNS and demyelination; however, these findings were confounded by spinal cord compression by virus-induced vertebral bone tumors. Here, we review current literature regarding animal models of PML, focusing on current trends in antiviral T cell immunity in non-lymphoid organs, including the CNS. Advances in our understanding of polyomavirus lifecycles, viral and host determinants of persistent infection, and T cell-mediated immunity to viral infections in the CNS warrant revisiting polyomavirus CNS infection in the mouse as a bona fide animal model for JCV-PML.
Collapse
Affiliation(s)
- Elizabeth L Frost
- Immunology and Molecular Pathogenesis Graduate Program, Emory University , Atlanta, GA , USA
| | - Aron E Lukacher
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine , Hershey, PA , USA
| |
Collapse
|
142
|
Sano Y, Nakano Y, Omoto M, Takao M, Ikeda E, Oga A, Nakamichi K, Saijo M, Maoka T, Sano H, Kawai M, Kanda T. Rituximab-associated progressive multifocal leukoencephalopathy derived from non-Hodgkin lymphoma: neuropathological findings and results of mefloquine treatment. Intern Med 2015; 54:965-70. [PMID: 25876582 DOI: 10.2169/internalmedicine.54.2308] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A 66-year-old man with non-Hodgkin lymphoma (NHL) developed progressive multifocal leukoencephalopathy (PML) after undergoing chemotherapy including rituximab. Although the administration of mefloquine at a dose of 500 mg weekly temporarily led to a dramatic decrease in the copy number of JC Virus DNA in the cerebrospinal fluid, the patient's symptoms gradually worsened. The CD4(+) T count remained continuously low, at least until approximately five months after the last cycle of chemotherapy. A postmortem examination performed 10 months after the onset of PML disclosed a severe condition associated with rituximab-treated PML originating from NHL and a high mefloquine concentration in the brain. The accumulation of further data regarding mefloquine treatment in PML cases may help to elucidate the optimal dosage and time window for effectively treating PML.
Collapse
Affiliation(s)
- Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Abstract
Rheumatologic diseases encompass autoimmune and inflammatory disorders of the joints and soft tissues that often involve multiple organ systems, including the central and peripheral nervous systems. Common features include constitutional symptoms, arthralgia and arthritis, myalgia, and sicca symptoms. Neurological manifestations may present in patients with preexisting rheumatologic diagnoses, occur concurrently with systemic signs and symptoms, or precede systemic manifestations by months to years. Rheumatic disorders presenting as neurological syndromes may pose diagnostic challenges. Advances in immunosuppressive treatment of rheumatologic disease have expanded the treatment armamentarium. However, serious neurotoxic effects have been reported with both old and newer agents. Familiarity with neurological manifestations of rheumatologic diseases, diagnosis, and potential nervous system consequences of treatment is important for rapid diagnosis and appropriate intervention. This article briefly reviews the diverse neurological manifestations and key clinical features of rheumatic disorders and the potential neurological complications of agents commonly used for treatment.
Collapse
|
144
|
Abstract
Monoclonal antibodies have become an important treatment option for a number of serious conditions. Concerns have arisen about the potential association of these products with progressive multifocal leukoencephalopathy (PML). A list of monoclonal antibodies authorized for sale was derived from the Health Canada Drug Product Database. Case reports of PML after exposure to a monoclonal antibody authorized for use in Canada were retrieved by searching Canada Vigilance and WHO adverse event databases and through a Pub MED/Medline literature search. 182 adverse event case reports were retrieved (adalimumab -1 case, alemtuzumab-14, bevacizumab -3, cetuximab -1, efalizumab - 8, ibritumomab tiuxetan-5, infliximab-4, natalizumab-32, and rituximab-114). The Canadian Product Monographs for natalizumab and ritiximab contain box warnings for PML. A natalizumab registry has been established.
Collapse
|
145
|
Jouneau S, Kerjouan M, Briens E, Lenormand JP, Meunier C, Letheulle J, Chiforeanu D, Lainé-Caroff C, Desrues B, Delaval P. La protéinose alvéolaire pulmonaire. Rev Mal Respir 2014; 31:975-91. [DOI: 10.1016/j.rmr.2014.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 08/20/2014] [Indexed: 01/30/2023]
|
146
|
Karonitsch T, Aringer M. [Biologics in SLE]. Wien Med Wochenschr 2014; 165:40-5. [PMID: 25411010 DOI: 10.1007/s10354-014-0322-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 10/02/2014] [Indexed: 11/24/2022]
Abstract
Biologics have become indispensable in the last decade in the treatment of the more common rheumatic diseases. For treating systemic lupus erythematodes (SLE), B-cell depletion, albeit off-label, has been a well-accepted strategy in severe and refractory disease. Unexpectedly, however, the results of the first randomized controlled rituximab trials in SLE were negative. New trials with improved study protocols are ongoing, which should resolve this issue. In 2012, with the approval of belimumab, SLE finally entered the era of approved biological therapies. The anti-Blys/BAFF antibody belimumab showed prevention of SLE flares, glucocorticoid sparing, and significant improvement in the quality of life of SLE patients, in part by drastically reducing immune complex mediated fatigue. Positive reports on further targeting approaches give hope that additional biological agents will be available for SLE therapy soon.
Collapse
Affiliation(s)
- Thomas Karonitsch
- Klinische Abteilung für Rheumatologie, Universitätsklinik für Innere Medizin III, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich,
| | | |
Collapse
|
147
|
Serana F, Chiarini M, Sottini A, Bertoli D, Giustini V, Tessitore MV, Caimi L, Capra R, Imberti L. Immunological biomarkers identifying natalizumab-treated multiple sclerosis patients at risk of progressive multifocal leukoencephalopathy. J Neuroimmunol 2014; 277:6-12. [PMID: 25468273 DOI: 10.1016/j.jneuroim.2014.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 10/27/2014] [Accepted: 10/29/2014] [Indexed: 01/27/2023]
Abstract
Natalizumab-induced progressive multifocal leukoencephalopathy appears to be unleashed by complex interactions between viral and immunological host factors leading the latent form of JC virus to become pathogenic. Positive anti-JC virus antibody status, prior use of immunosuppressants, and increasing duration of natalizumab treatment have been proposed as risk factors for progressive multifocal leukoencephalopathy in multiple sclerosis patients, but while they may help to identify the most appropriate patients for natalizumab, their use have some limitations. Therefore, a large body of studies is ongoing to identify alternative, reliable immunological markers capable to improve the safety and efficacy of therapy, and to guide tailored clinical decisions.
Collapse
Affiliation(s)
- Federico Serana
- Centro di Ricerca Emato-oncologica AIL (CREA), Diagnostics Department, Spedali Civili of Brescia, Italy
| | - Marco Chiarini
- Centro di Ricerca Emato-oncologica AIL (CREA), Diagnostics Department, Spedali Civili of Brescia, Italy
| | - Alessandra Sottini
- Centro di Ricerca Emato-oncologica AIL (CREA), Diagnostics Department, Spedali Civili of Brescia, Italy
| | - Diego Bertoli
- Centro di Ricerca Emato-oncologica AIL (CREA), Diagnostics Department, Spedali Civili of Brescia, Italy
| | - Viviana Giustini
- Centro di Ricerca Emato-oncologica AIL (CREA), Diagnostics Department, Spedali Civili of Brescia, Italy
| | - Marion Vaglio Tessitore
- Centro di Ricerca Emato-oncologica AIL (CREA), Diagnostics Department, Spedali Civili of Brescia, Italy
| | - Luigi Caimi
- Centro di Ricerca Emato-oncologica AIL (CREA), Diagnostics Department, Spedali Civili of Brescia, Italy
| | - Ruggero Capra
- Multiple Sclerosis Center, Spedali Civili of Brescia, Italy
| | - Luisa Imberti
- Centro di Ricerca Emato-oncologica AIL (CREA), Diagnostics Department, Spedali Civili of Brescia, Italy.
| |
Collapse
|
148
|
Tuccori M, Montagnani S, Capogrosso-Sansone A, Mantarro S, Antonioli L, Fornai M, Blandizzi C. Adverse reactions to oncologic drugs: spontaneous reporting and signal detection. Expert Rev Clin Pharmacol 2014; 8:61-75. [PMID: 25363790 DOI: 10.1586/17512433.2015.974555] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oncology is one of the areas of medicine with the most active research being conducted on new drugs. New pharmacological entities frequently enter the clinical arena, and therefore, the safety profile of anticancer products deserves continuous monitoring. However, only very severe and (unusual) suspected adverse drug reactions (ADRs) are usually reported, since cancer patients develop ADRs very frequently and some practical selectivity must be used. Notably, a recent study was able to identify 76 serious ADRs reported in updated drug labels of oncologic drugs and 50% of them (n = 38) were potentially fatal. Of these, 49 and 58%, respectively, were not described in initial drug labels. The aims of this article are to provide an overview about spontaneous reporting of ADRs of oncologic drugs and to discuss the available methods to analyze the safety of anticancer drugs using databases of spontaneous ADR reporting.
Collapse
Affiliation(s)
- Marco Tuccori
- Tuscan Regional Centre of Pharmacovigilance, Pisa, Italy
| | | | | | | | | | | | | |
Collapse
|
149
|
Verma NK, Kelleher D. Adaptor regulation of LFA-1 signaling in T lymphocyte migration: Potential druggable targets for immunotherapies? Eur J Immunol 2014; 44:3484-99. [PMID: 25251823 DOI: 10.1002/eji.201344428] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 09/16/2014] [Accepted: 09/22/2014] [Indexed: 01/24/2023]
Abstract
The integrin lymphocyte function associated antigen-1 (LFA-1) plays a key role in leukocyte trafficking and in adaptive immune responses through interactions with adhesive ligands, such as ICAM-1. Specific blockade of these interactions has validated LFA-1 as a therapeutic target in many chronic inflammatory diseases, however LFA-1 antagonists have not been clinically successful due to the development of a general immunosuppression, causing fatal side effects. Growing evidence has now established that LFA-1 mediates an array of intracellular signaling pathways by triggering a number of downstream molecules. In this context, a class of multimodular domain-containing proteins capable of recruiting two or more effector molecules, collectively known as "adaptor proteins," has emerged as important mediators in LFA-1 signal transduction. Here, we provide an overview of the adaptor proteins involved in the intracellular signaling cascades by which LFA-1 regulates T-cell motility and immune responses. The complexity of the LFA-1-associated signaling delineated in this review suggests that it may be an important and challenging focus for future research, enabling the identification of "tunable" targets for the development of immunotherapies.
Collapse
Affiliation(s)
- Navin K Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Singapore Eye Research Institute, Singapore, Singapore
| | | |
Collapse
|
150
|
Schwab N, Schneider-Hohendorf T, Wiendl H. Therapeutic uses of anti-α4-integrin (anti-VLA-4) antibodies in multiple sclerosis. Int Immunol 2014; 27:47-53. [PMID: 25326459 DOI: 10.1093/intimm/dxu096] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis (MS) is a disorder of putative autoimmune origin, where immune cells invade the central nervous system and cause damage by attacking the myelin sheath of nerve cells. The blockade of the integrin very late antigen-4 (VLA-4) with the monoclonal antibody natalizumab has become the most effective therapy against MS since its approval in 2004. It is assumed that the inhibition of VLA-4-mediated immune cell adhesion to the endothelium of the blood-brain barrier (BBB) alleviates pathogenic processes of MS and, therefore, reduces disease severity and burden. Not all approaches to treat additional immune-mediated disorders (e.g. Rasmussen encephalitis and neuromyelitis optica) with natalizumab have been successful, but allowed researchers to gain additional insight into mechanisms of specific immune cell subsets' migration through the BBB in the human system. While the long-term efficacy and general tolerability of natalizumab in MS are clear, the over 400 cases of natalizumab-associated progressive multifocal leukoencephalopathy (PML) have been of great concern and methods of risk stratification in patients have become a major area of research. Modern risk stratification includes established factors such as treatment duration, previous immune-suppressive therapy, and anti-John Cunningham virus (JCV) antibody seropositivity, but also experimental factors such as anti-JCV antibody titers and levels of L-selectin. Today, anti-VLA-4 therapy is reserved for patients with highly active relapsing-remitting MS and patients are monitored closely for early signs of potential PML.
Collapse
Affiliation(s)
- Nicholas Schwab
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | | | - Heinz Wiendl
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| |
Collapse
|