1
|
Manoutcharian K, Gevorkian G. Recombinant Antibody Fragments for Neurological Disorders: An Update. Curr Neuropharmacol 2024; 22:2157-2167. [PMID: 37646225 PMCID: PMC11337690 DOI: 10.2174/1570159x21666230830142554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/15/2023] [Accepted: 07/13/2023] [Indexed: 09/01/2023] Open
Abstract
Recombinant antibody fragments are promising alternatives to full-length immunoglobulins, creating big opportunities for the pharmaceutical industry. Nowadays, antibody fragments such as antigen-binding fragments (Fab), single-chain fragment variable (scFv), single-domain antibodies (sdAbs), and bispecific antibodies (bsAbs) are being evaluated as diagnostics or therapeutics in preclinical models and in clinical trials. Immunotherapy approaches, including passive transfer of protective antibodies, have shown therapeutic efficacy in several animal models of Alzheimer's disease (AD), Parkinson's disease (PD), frontotemporal dementia (FTD), Huntington's disease (HD), transmissible spongiform encephalopathies (TSEs) and multiple sclerosis (MS). There are various antibodies approved by the Food and Drug Administration (FDA) for treating multiple sclerosis and two amyloid beta-specific humanized antibodies, Aducanumab and Lecanemab, for AD. Our previous review summarized data on recombinant antibodies evaluated in pre-clinical models for immunotherapy of neurodegenerative diseases. Here, we explore recent studies in this fascinating research field, give an update on new preventive and therapeutic applications of recombinant antibody fragments for neurological disorders and discuss the potential of antibody fragments for developing novel approaches for crossing the blood-brain barrier (BBB) and targeting cells and molecules of interest in the brain.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), CDMX, Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), CDMX, Mexico
| |
Collapse
|
2
|
Manchon E, Laplaud D, Vukusic S, Labauge P, Moreau T, Kobelt G, Grouin JM, Lotz M, Pau D, Christine LF. Efficacy, safety and patient reported outcomes in patients with active relapsing multiple sclerosis treated with ocrelizumab: Final results from the PRO-MSACTIVE study. Mult Scler Relat Disord 2022; 68:104109. [PMID: 36007299 DOI: 10.1016/j.msard.2022.104109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/03/2022] [Accepted: 08/12/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Ocrelizumab, a humanized anti-CD20 monoclonal antibody, has been approved in Europe for the treatment of adult patients with active relapsing multiple sclerosis (RMS) and primary progressive multiple sclerosis (PPMS), on the basis of previous phase III studies. However, limited data were available on ocrelizumab efficacy in RMS according to the Lublin definition of activity (clinical and/or imaging features) used in the current drug label. The PRO-MSACTIVE study was thus designed to provide additional data on ocrelizumab efficacy according to this definition, and also on safety and patient reported outcomes (PROs). METHODS PRO-MSACTIVE is a national, multicenter, open-label, single-arm phase IV French study, conducted in patients with active RMS (relapsing-remitting multiple sclerosis, RRMS, or secondary progressive multiple sclerosis, SPMS). The primary endpoint, which was assessed at week (W) 48, was defined as the proportion of patients free of disease activity (defined by no relapses and no T1 gadolinium-enhancing nor new and/or enlarging T2 lesions using brain MRI). Disease activity, disability and PROs using 6 questionnaires for disease severity, quality of life, impact on work productivity, and treatment satisfaction were described at W24 and W48. Adverse events were described until W72. RESULTS Among the 422 analyzed patients (RRMS: 376, SPMS: 46), 63.3% (95% CI [58.5%; 67.9%]) were free of disease activity at W48 (RRMS: 62.2% [57.1%; 67.2%], SPMS: 71.7% [56.5%; 84.0%]). A total of 358 patients (84.8%; RRMS: 84.6%, SPMS: 87.0%) were relapse-free up to W48, and the overall adjusted annualized relapse rate was 0.14 (RRMS: 0.15, SPMS: 0.09). Overall, 67.8% of patients (RRMS: 66.8%, SPMS: 76.1%) had no evidence of MRI activity (no T1 gadolinium-enhancing lesions [83.4%] and no new/enlarging T2 lesions [75.1%]); 58.5% of patients (RRMS: 57.7%, SPMS: 65.2%) achieved No Evidence of Disease Activity (NEDA: no relapses, no confirmed disability progression, and no MRI activity) at W48. All PRO scores were stable between the first dose of ocrelizumab and W48 and better outcomes were seen for patients having an EDSS score ≥4. Overall, 89.3% of patients reported adverse events, 62.3% adverse events assessed as related to ocrelizumab, and 8.5% serious adverse events. No serious infusion-related reactions, opportunistic infections, progressive multifocal leukoencephalopathy, nor deaths were reported. No new safety signal was identified. CONCLUSION These data confirm the efficacy of ocrelizumab in a pragmatic setting and its favorable benefit-risk profile in patients with RMS. (ClinicalTrials.gov identifier: NCT03589105; EudraCT identifier: 2018-000780-91).
Collapse
Affiliation(s)
- Eric Manchon
- Centre Hospitalier de Gonesse, Service de Neurologie, Gonesse, France.
| | - David Laplaud
- Nantes Université, Service de Neurologie, Centre Hospitalier Universitaire de Nantes, CIC INSERM 1413, Center for Research in Transplantation and Translational Immunology, INSERM UMR 1064, Nantes, France
| | - Sandra Vukusic
- Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, Service de Neurologie, Centre de Recherche en Neurosciences de Lyon, OFSEP, INSERM 1028 et CNRS UMR 5292, Université Claude Bernard de Lyon, Eugène Devic EDMUS Foundation, Bron, France
| | - Pierre Labauge
- Centre Hospitalier Universitaire de Montpellier, Hôpital Gui de Chauliac, Service de Neurologie, Montpellier, France
| | - Thibault Moreau
- Centre Hospitalier Universitaire de Dijon Bourgogne, Hôpital François Mitterrand, Maladies Inflammatoires du Système Nerveux et Neurologie Générale, Service de Neurologie, Dijon, France
| | | | | | | | - David Pau
- Roche SAS, Boulogne-Billancourt, France
| | - Lebrun Frenay Christine
- Centre Hospitalier Universitaire Pasteur 2, Service de Neurologie, CRCSEP, Unitéde Recherche Clinique Côte d'Azur (UR2CA-URRIS), Nice, France
| |
Collapse
|
3
|
Krämer J, Wiendl H. What Have Failed, Interrupted, and Withdrawn Antibody Therapies in Multiple Sclerosis Taught Us? Neurotherapeutics 2022; 19:785-807. [PMID: 35794296 PMCID: PMC9294122 DOI: 10.1007/s13311-022-01246-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2022] [Indexed: 12/13/2022] Open
Abstract
In the past two decades, monoclonal antibodies (mAbs) have revolutionized the treatment of multiple sclerosis (MS). However, a remarkable number of mAbs failed due to negative study results were withdrawn because of unexpected serious adverse events (SAEs) or due to studies being halted for other reasons. While trials with positive outcomes are usually published in prestigious journals, negative trials are merely published as abstracts or not at all. This review summarizes MS mAbs that have either failed in phase II-III trials, have been interrupted for various reasons, or withdrawn from the market since 2015. The main conclusions that can be drawn from these 'negative' experiences are as follows. mAbs that have been proven to be safe in other autoimmune conditions, will not have the same safety profile in MS due to immunopathogenetic differences in these diseases (e.g., daclizumab). Identification of SAEs in clinical trials is difficult highlighting the importance of phase IV studies. Memory B cells are central players in MS immunopathogenesis (e.g., tabalumab). The pathophysiological mechanisms of disease progression are independent of leukocyte 'outside-in' traffic which drives relapses in MS. Therefore, therapies for progressive MS must be able to sufficiently cross the blood-brain barrier. Sufficiently long trial duration and multicomponent outcome measures are important for clinical studies in progressive MS. The success of trials on remyelination-promoting therapies mainly depends on the sufficient high dose of mAb, the optimal readout for 'proof of concept', time of treatment initiation, and appropriate selection of patients. Failed strategies are highly important to better understand assumed immunopathophysiological mechanisms and optimizing future trial designs.
Collapse
Affiliation(s)
- Julia Krämer
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| | - Heinz Wiendl
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Muenster, Germany
| |
Collapse
|
4
|
Agoston DV. COVID-19 and Traumatic Brain Injury (TBI); What We Can Learn From the Viral Pandemic to Better Understand the Biology of TBI, Improve Diagnostics and Develop Evidence-Based Treatments. Front Neurol 2021; 12:752937. [PMID: 34987462 PMCID: PMC8720751 DOI: 10.3389/fneur.2021.752937] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/01/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Denes V. Agoston
- Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
5
|
Improved antibody adsorption performance of phenyl-based mixed-mode adsorbents by adjusting the functional group of ligand. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2021.108092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
6
|
Savizi ISP, Motamedian E, E Lewis N, Jimenez Del Val I, Shojaosadati SA. An integrated modular framework for modeling the effect of ammonium on the sialylation process of monoclonal antibodies produced by CHO cells. Biotechnol J 2021; 16:e2100019. [PMID: 34021707 DOI: 10.1002/biot.202100019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Monoclonal antibodies (mABs) have emerged as one of the most important therapeutic recombinant proteins in the pharmaceutical industry. Their immunogenicity and therapeutic efficacy are influenced by post-translational modifications, specifically the glycosylation process. Bioprocess conditions can influence the intracellular process of glycosylation. Among all the process conditions that have been recognized to affect the mAB glycoforms, the detailed mechanism underlying how ammonium could perturb glycosylation remains to be fully understood. It was shown that ammonium induces heterogeneity in protein glycosylation by altering the sialic acid content of glycoproteins. Hence, understanding this mechanism would aid pharmaceutical manufacturers to ensure consistent protein glycosylation. METHODS Three different mechanisms have been proposed to explain how ammonium influences the sialylation process. In the first, the inhibition of CMP-sialic acid transporter, which transports CMP-sialic acid (sialylation substrate) into the Golgi, by an increase in UDP-GlcNAc content that is brought about by the augmented incorporation of ammonium into glucosamine formation. In the second, ammonia diffuses into the Golgi and raises its pH, thereby decreasing the sialyltransferase enzyme activity. In the third, the reduction of sialyltransferase enzyme expression level in the presence of ammonium. We employed these mechanisms in a novel integrated modular platform to link dynamic alteration in mAB sialylation process with extracellular ammonium concentration to elucidate how ammonium alters the sialic acid content of glycoproteins. RESULTS Our results show that the sialylation reaction rate is insensitive to the first mechanism. At low ammonium concentration, the second mechanism is the controlling mechanism in mAB sialylation and by increasing the ammonium level (< 8 mM) the third mechanism becomes the controlling mechanism. At higher ammonium concentrations (> 8 mM) the second mechanism becomes predominant again. CONCLUSION The presented model in this study provides a connection between extracellular ammonium and the monoclonal antibody sialylation process. This computational tool could help scientists to develop and formulate cell culture media. The model illustrated here can assist the researchers to select culture media that ensure consistent mAB sialylation.
Collapse
Affiliation(s)
- Iman Shahidi Pour Savizi
- Faculty of Chemical Engineering, Biotechnology Department, Tarbiat Modares University, Tehran, Iran
| | - Ehsan Motamedian
- Faculty of Chemical Engineering, Biotechnology Department, Tarbiat Modares University, Tehran, Iran
| | - Nathan E Lewis
- Department of Bioengineering, University of California, La Jolla, California, USA.,School of Medicine, Novo Nordisk Foundation Center for Biosustainability at the University of California, La Jolla, California, USA.,Department of Pediatrics, School of Medicine, University of California, La Jolla, California, USA
| | | | - Seyed Abbas Shojaosadati
- Faculty of Chemical Engineering, Biotechnology Department, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
7
|
Mark VW. Retention of Physical Gains in the Community Following Physical Training for Multiple Sclerosis: A Systematic Review and Implications. Semin Neurol 2021; 41:177-188. [PMID: 33690875 DOI: 10.1055/s-0041-1725139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Multiple sclerosis (MS) is a progressive neurological illness whose typically young adult onset results in a nearly entire lifetime of worsening disability. But despite being an unrelenting neurodegenerative disease, numerous clinical trials over the past 40 years for MS have vigorously attempted to improve or at least stabilize declining physical function. Although the vast majority of the studies assessed training effects only within controlled laboratory or clinic settings, in recent years a growing interest has emerged to test whether newer therapies can instead benefit real-life activities in the community. Nonetheless, comparatively little attention has been paid to whether the training gains can be retained for meaningful periods. This review discusses the comparative success of various physical training methods to benefit within-community activities in MS, and whether the gains can be retained long afterward. This review will suggest future research directions toward establishing efficacious treatments that can allow persons with MS to reclaim their physical abilities and maximize functionality for meaningful periods.
Collapse
Affiliation(s)
- Victor W Mark
- Departments of Physical Medicine and Rehabilitation, Neurology, and Psychology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
8
|
Tarlinton RE, Martynova E, Rizvanov AA, Khaiboullina S, Verma S. Role of Viruses in the Pathogenesis of Multiple Sclerosis. Viruses 2020; 12:E643. [PMID: 32545816 PMCID: PMC7354629 DOI: 10.3390/v12060643] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/07/2020] [Accepted: 06/10/2020] [Indexed: 12/17/2022] Open
Abstract
Multiple sclerosis (MS) is an immune inflammatory disease, where the underlying etiological cause remains elusive. Multiple triggering factors have been suggested, including environmental, genetic and gender components. However, underlying infectious triggers to the disease are also suspected. There is an increasing abundance of evidence supporting a viral etiology to MS, including the efficacy of interferon therapy and over-detection of viral antibodies and nucleic acids when compared with healthy patients. Several viruses have been proposed as potential triggering agents, including Epstein-Barr virus, human herpesvirus 6, varicella-zoster virus, cytomegalovirus, John Cunningham virus and human endogenous retroviruses. These viruses are all near ubiquitous and have a high prevalence in adult populations (or in the case of the retroviruses are actually part of the genome). They can establish lifelong infections with periods of reactivation, which may be linked to the relapsing nature of MS. In this review, the evidence for a role for viral infection in MS will be discussed with an emphasis on immune system activation related to MS disease pathogenesis.
Collapse
Affiliation(s)
- Rachael E. Tarlinton
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough LE12 5RD, UK;
| | - Ekaterina Martynova
- Insititute of Fundamental Medicine and Biology Kazan Federal University, 420008 Kazan, Russia; (E.M.); (A.A.R.)
| | - Albert A. Rizvanov
- Insititute of Fundamental Medicine and Biology Kazan Federal University, 420008 Kazan, Russia; (E.M.); (A.A.R.)
| | | | - Subhash Verma
- School of Medicine, University of Nevada, Reno, NV 89557, USA;
| |
Collapse
|
9
|
Metaxakis A, Petratou D, Tavernarakis N. Molecular Interventions towards Multiple Sclerosis Treatment. Brain Sci 2020; 10:brainsci10050299. [PMID: 32429225 PMCID: PMC7287961 DOI: 10.3390/brainsci10050299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune life-threatening disease, afflicting millions of people worldwide. Although the disease is non-curable, considerable therapeutic advances have been achieved through molecular immunotherapeutic approaches, such as peptides vaccination, administration of monoclonal antibodies, and immunogenic copolymers. The main aims of these therapeutic strategies are to shift the MS-related autoimmune response towards a non-inflammatory T helper 2 (Th2) cells response, inactivate or ameliorate cytotoxic autoreactive T cells, induce secretion of anti-inflammatory cytokines, and inhibit recruitment of autoreactive lymphocytes to the central nervous system (CNS). These approaches can efficiently treat autoimmune encephalomyelitis (EAE), an essential system to study MS in animals, but they can only partially inhibit disease progress in humans. Nevertheless, modern immunotherapeutic techniques remain the most promising tools for the development of safe MS treatments, specifically targeting the cellular factors that trigger the initiation of the disease.
Collapse
Affiliation(s)
- Athanasios Metaxakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Nikolaou Plastira 100, 70013 Heraklion, Greece; (A.M.); (D.P.)
| | - Dionysia Petratou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Nikolaou Plastira 100, 70013 Heraklion, Greece; (A.M.); (D.P.)
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Nikolaou Plastira 100, 70013 Heraklion, Greece; (A.M.); (D.P.)
- Department of Basic Sciences, Faculty of Medicine, University of Crete, 71110 Heraklion, Greece
- Correspondence: ; Tel.: +30-2810-391066
| |
Collapse
|
10
|
Kim W, Kim HJ. Monoclonal Antibody Therapies for Multiple Sclerosis and Neuromyelitis Optica Spectrum Disorder. J Clin Neurol 2020; 16:355-368. [PMID: 32657055 PMCID: PMC7354979 DOI: 10.3988/jcn.2020.16.3.355] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/01/2019] [Accepted: 12/02/2019] [Indexed: 12/21/2022] Open
Abstract
Considerable progress has been made in treatments for multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) over the last several decades. However, the present treatments do not show satisfactory efficacy or safety in a considerable proportion of patients, who experience relapse or disability progression despite receiving treatment and suffer from side effects, which can be severe. Improvements in the understanding of the pathophysiologies of MS and NMOSD have led to numerous therapeutic approaches being proposed and developed. Monoclonal antibodies (mAbs) are receiving increasing attention because of their specificity of action and likelihood of high efficacy with fewer side effects. Many mAbs have been evaluated, and some have been approved for MS or NMOSD treatment. This article reviews the use of mAbs for treating MS and NMOSD, including summarizing their mechanisms of action, efficacy, and safety profiles.
Collapse
Affiliation(s)
- Woojun Kim
- Department of Neurology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho Jin Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, Korea.
| |
Collapse
|
11
|
Monoclonal Antibodies in Multiple Sclerosis: Present and Future. Biomedicines 2019; 7:biomedicines7010020. [PMID: 30875812 PMCID: PMC6466331 DOI: 10.3390/biomedicines7010020] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 01/09/2023] Open
Abstract
The global incidence of multiple sclerosis (MS) appears to be increasing. Although it may not be associated with a high mortality rate, this disease has a high morbidity rate which affects the quality of life of patients and reduces their ability to do their activities of daily living. Thankfully, the development of novel disease modifying therapies continues to increase. Monoclonal antibodies (MABs) have become a mainstay of MS treatment and they are likely to continue to be developed for the treatment of this disease. Specifically, MABs have proven to be some of the most efficacious treatments at reducing relapses and the inflammation in MS patients, including the first treatment for primary progressive MS and are being explored as reparative/remyelinating agents as well. These relatively new treatments will be reviewed here to help evaluate their efficacy, adverse events, immunogenicity, and benefit-risk ratios in the treatment of the diverse spectrum of MS. The focus will be on MABs that are currently approved or may be approved in the near future.
Collapse
|
12
|
Pisaturo M, Di Caprio G, Calò F, Portunato F, Martini S, Coppola N. Management of HBV reactivation in non-oncological patients. Expert Rev Anti Infect Ther 2019; 16:611-624. [PMID: 30058401 DOI: 10.1080/14787210.2018.1505501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION HBV reactivation (HBVr) in patients undergoing immunosuppressive therapy is a well-known event. While there are clear directives on the management of current or resolved HBV infection in onco-hematological diseases, there are few data regarding patients with non-oncological diseases. Thus, the aim of the present review is to evaluate HBVr in patients with non-oncological diseases, and identify the management of these patients to prevent HBVr. Areas covered: Original papers, case reports and meta-analyses reporting data on HBVr of current or resolved infection in gastrointestinal, dermatological, rheumatologic and neurological diseases were evaluated. Expert commentary: In HBsAg-positive subjects, those with HBV-related hepatitis (both HBeAg-positive or negative) should be treated with a high genetic barrier nucleos(t)ide analog. The patients with HBV-infection (both HBeAg-positive and negative) an antiviral prophylaxis should be used, with lamivudine in those HBeAg-negative without signs of advanced liver disease, and with ETV, TDF or TAF in all the HBeAg-positive or in those HBeAg-negative with signs of advanced liver disease. In HBsAg-negative/anti-HBc positive subjects, when the risk of HBV reactivation is moderate (use of B-cell depleting agents), a prophylaxis-strategy may be considered; instead, in those with low risk of HBVr, a pre-emptive therapy strategy may be used.
Collapse
Affiliation(s)
- Mariantonietta Pisaturo
- a Department of Mental Health and Public Medicine, Section of Infectious Diseases , University of Campania , Naples , Italy
| | - Giovanni Di Caprio
- a Department of Mental Health and Public Medicine, Section of Infectious Diseases , University of Campania , Naples , Italy
| | - Federica Calò
- a Department of Mental Health and Public Medicine, Section of Infectious Diseases , University of Campania , Naples , Italy
| | - Federica Portunato
- a Department of Mental Health and Public Medicine, Section of Infectious Diseases , University of Campania , Naples , Italy
| | - Salvatore Martini
- a Department of Mental Health and Public Medicine, Section of Infectious Diseases , University of Campania , Naples , Italy.,b Section of Infectio us Diseases , A.O.R.N. Dei Colli, Cotugno Hospital , Napoli , Italy
| | - Nicola Coppola
- a Department of Mental Health and Public Medicine, Section of Infectious Diseases , University of Campania , Naples , Italy.,c Section of Infectious Diseases , A.O.R.N S.Anna S. Sebastiano Caserta , Caserta , Italy
| |
Collapse
|
13
|
Kumar NN, Pizzo ME, Nehra G, Wilken-Resman B, Boroumand S, Thorne RG. Passive Immunotherapies for Central Nervous System Disorders: Current Delivery Challenges and New Approaches. Bioconjug Chem 2018; 29:3937-3966. [PMID: 30265523 PMCID: PMC7234797 DOI: 10.1021/acs.bioconjchem.8b00548] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Passive immunotherapy, i.e., the administration of exogenous antibodies that recognize a specific target antigen, has gained significant momentum as a potential treatment strategy for several central nervous system (CNS) disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and brain cancer, among others. Advances in antibody engineering to create therapeutic antibody fragments or antibody conjugates have introduced new strategies that may also be applied to treat CNS disorders. However, drug delivery to the CNS for antibodies and other macromolecules has thus far proven challenging, due in large part to the blood-brain barrier and blood-cerebrospinal fluid barriers that greatly restrict transport of peripherally administered molecules from the systemic circulation into the CNS. Here, we summarize the various passive immunotherapy approaches under study for the treatment of CNS disorders, with a primary focus on disease-specific and target site-specific challenges to drug delivery and new, cutting edge methods.
Collapse
Affiliation(s)
- Niyanta N. Kumar
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Michelle E. Pizzo
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
- Clinical Neuroengineering Training Program, University of
Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Geetika Nehra
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Brynna Wilken-Resman
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Sam Boroumand
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Robert G. Thorne
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
- Clinical Neuroengineering Training Program, University of
Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Neuroscience Training Program & Center for
Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin 53705, United
States
- Cellular and Molecular Pathology Graduate Training Program,
University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
14
|
Vala M, Jordan LR, Warrington AE, Maher LJ, Rodriguez M, Wittenberg NJ, Oh SH. Surface Plasmon Resonance Sensing on Naturally Derived Membranes: A Remyelination-Promoting Human Antibody Binds Myelin with Extraordinary Affinity. Anal Chem 2018; 90:12567-12573. [DOI: 10.1021/acs.analchem.8b02664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Milan Vala
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Luke R. Jordan
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Arthur E. Warrington
- Departments of Neurology and Immunology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - L. James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Moses Rodriguez
- Departments of Neurology and Immunology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Nathan J. Wittenberg
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Sang-Hyun Oh
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
15
|
Manoutcharian K, Perez-Garmendia R, Gevorkian G. Recombinant Antibody Fragments for Neurodegenerative Diseases. Curr Neuropharmacol 2018; 15:779-788. [PMID: 27697033 PMCID: PMC5771054 DOI: 10.2174/1570159x01666160930121647] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/04/2016] [Accepted: 09/28/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Recombinant antibody fragments are promising alternatives to full-length immunoglobulins and offer important advantages compared with conventional monoclonal antibodies: extreme specificity, higher affinity, superior stability and solubility, reduced immunogenicity as well as easy and inexpensive large-scale production. OBJECTIVE In this article we will review and discuss recombinant antibodies that are being evaluated for neurodegenerative diseases in pre-clinical models and in clinical studies and will summarize new strategies that are being developed to optimize their stability, specificity and potency for advancing their use. METHODS Articles describing recombinant antibody fragments used for neurological diseases were selected (PubMed) and evaluated for their significance. RESULTS Different antibody formats such as single-chain fragment variable (scFv), single-domain antibody fragments (VHHs or sdAbs), bispecific antibodies (bsAbs), intrabodies and nanobodies, are currently being studied in pre-clinical models of cancer as well as infectious and autoimmune diseases and many of them are being tested as therapeutics in clinical trials. Immunotherapy approaches have shown therapeutic efficacy in several animal models of Alzheimer´s disease (AD), Parkinson disease (PD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD), Huntington disease (HD), transmissible spongiform encephalopathies (TSEs) and multiple sclerosis (MS). It has been demonstrated that recombinant antibody fragments may neutralize toxic extra- and intracellular misfolded proteins involved in the pathogenesis of AD, PD, DLB, FTD, HD or TSEs and may target toxic immune cells participating in the pathogenesis of MS. CONCLUSION Recombinant antibody fragments represent a promising tool for the development of antibody-based immunotherapeutics for neurodegenerative diseases.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Mexico DF. Mexico
| | - Roxanna Perez-Garmendia
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Mexico DF. Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP 04510, Mexico. 0
| |
Collapse
|
16
|
Up-to-date knowledge about the association between multiple sclerosis and the reactivation of human endogenous retrovirus infections. J Neurol 2018; 265:1733-1739. [DOI: 10.1007/s00415-018-8783-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022]
|
17
|
Hosseini A, Estiri H, Akhavan Niaki H, Alizadeh A, Abdolhossein Zadeh B, Ghaderian SMH, Farjadfar A, Fallah A. Multiple Sclerosis Gene Therapy with Recombinant Viral Vectors: Overexpression of IL-4, Leukemia Inhibitory Factor, and IL-10 in Wharton's Jelly Stem Cells Used in EAE Mice Model. CELL JOURNAL 2017; 19:361-374. [PMID: 28836399 PMCID: PMC5570402 DOI: 10.22074/cellj.2017.4497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 09/07/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Immunotherapy and gene therapy play important roles in modern medicine. The aim of this study is to evaluate the overexpression of interleukin-4 (IL-4), IL-10 and leukemia inhibitory factor (LIF) in Wharton's jelly stem cells (WJSCs) in the experimental autoimmune encephalomyelitis (EAE) mice model. MATERIALS AND METHODS In this experimental study, a DNA construction containing IL- 4, IL-10 and LIF was assembled to make a polycistronic vector (as the transfer vector). Transfer and control vectors were co-transfected into Human Embryonic Kidney 293 (HEK-293T) cells with helper plasmids which produced recombinant lentiviral viruses (rLV). WJSCs were transduced with rLV to make recombinant WJSC (rWJSC). In vitro protein and mRNA overexpression of IL-4, LIF, and IL-10 were evaluated using quantitative polymerase chain reaction (qPCR), enzyme-linked immunosorbent assay (ELISA) and western blot (WB) analysis. EAE was induced in mice by MOG-CFA and pertussis toxin. EAE mice were injected twice with 2×105 rWJSCs. The in vivo level of IL-4, LIF, IL-10 cytokines and IL-17 were measured by ELISA. Brain tissues were analyzed histologically for evaluation of EAE lesions. RESULTS Isolated WJSCs were performed to characterize by in vitro differentiation and surface markers were analyzed by flow cytometry method. Cloning of a single lentiviral vector with five genes was done successfully. Transfection of transfer and control vectors were processed based on CaPO4 method with >90% efficiency. Recombinant viruses were produced and results of titration showed 2-3×107 infection-unit/ml. WJSCs were transduced using recombinant viruses. IL-4, IL-10 and LIF overexpression were confirmed by ELISA, WB and qPCR. The EAE mice treated with rWJSC showed reduction of Il-17, and brain lesions as well as brain cellular infiltration, in vivo. Weights and physical activity were improved in gene-treated group. CONCLUSIONS These results showed that gene therapy using anti-inflammatory cytokines can be a promising approach against multiple sclerosis (MS). In addition, considering the immunomodulatory potential of WJSCs, an approach using a combination of WJSCs and gene therapy will enhance the treatment efficacy.
Collapse
Affiliation(s)
- Ahmad Hosseini
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Cell Biology and Anatomical Science, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hajar Estiri
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Iranian Institute of Cell and Gene Therapy, Tehran, Iran
| | - Haleh Akhavan Niaki
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Babol, Iran.,Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Akram Alizadeh
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Baharak Abdolhossein Zadeh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Akbar Farjadfar
- Department of Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Ali Fallah
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,BioViva USA Inc, Bainbridge Island WA, USA
| |
Collapse
|
18
|
Neuteboom R, Wilbur C, Van Pelt D, Rodriguez M, Yeh A. The Spectrum of Inflammatory Acquired Demyelinating Syndromes in Children. Semin Pediatr Neurol 2017; 24:189-200. [PMID: 29103426 DOI: 10.1016/j.spen.2017.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Acquired demyelinating syndromes in childhood comprise a spectrum of monophasic and recurrent inflammatory conditions of the central nervous system. Examples of monophasic conditions include, clinically isolated syndromes such as optic neuritis and transverse myelitis, as well as acute disseminated encephalomyelitis, whereas recurrent disorders include entities such as multiple sclerosis and neuromyelitis optica spectrum disorder. Knowledge about these disorders has expanded due to rigorously evaluated diagnostic criteria, magnetic resonance imaging features, outcomes, and serum biomarkers in these disorders. This review aims to provide a summary of clinical developments in pediatric acquired demyelinating syndromes, with a special focus on diagnostic criteria, neuroinflammatory markers, burden of disease in addition to current and potential future treatment options.
Collapse
Affiliation(s)
- Rinze Neuteboom
- Department of Pediatric Neurology, Dutch Pediatric MS Center, Sophia's Children's Hospital, Erasmus MC, Rotterdam, The Netherlands.
| | - Colin Wilbur
- Department of Pediatric Neurology, Sick Kids Children's Hospital, Toronto, Canada
| | | | | | - Ann Yeh
- Department of Pediatric Neurology, Sick Kids Children's Hospital, Toronto, Canada
| |
Collapse
|