1
|
Neunie OAM, Rabbani W, Baker D, Chambers ES, Pfeffer PE, Kang AS. Immunogenicity of biologics used in the treatment of asthma. Hum Antibodies 2024; 32:121-128. [PMID: 38905039 DOI: 10.3233/hab-240002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
OBJECTIVE Asthma is a major global disease affecting adults and children, which can lead to hospitalization and death due to breathing difficulties. Although targeted monoclonal antibody therapies have revolutionized treatment of severe asthma, some patients still fail to respond. Here we critically evaluate the literature on biologic therapy failure in asthma patients with particular reference to anti-drug antibody production, and subsequent loss of response, as the potential primary cause of drug failure in asthma patients. RECENT FINDINGS Encouragingly, asthma in most cases responds to treatment, including the use of an increasing number of biologic drugs in moderate to severe disease. This includes monoclonal antibody inhibitors of immunoglobulin E and cytokines, including interleukin 4, 5, or 13 and thymic stromal lymphopoietin. These limit mast cell and eosinophil activity that cause the symptomatic small airways obstruction and exacerbations. SUMMARY Despite humanization of the antibodies, it is evident that benralizumab; dupilumab; mepolizumab; omalizumab; reslizumab and tezepelumab all induce anti-drug antibodies to some extent. These can contribute to adverse events including infusion reactions, serum sickness, anaphylaxis and potentially disease activity due to loss of therapeutic function. Monitoring anti-drug antibodies (ADA) may allow prediction of future treatment-failure in some individuals allowing treatment cessation and switching therefore potentially limiting disease breakthrough.
Collapse
Affiliation(s)
- Omario A M Neunie
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Oral Immunobiology and Regenerative Medicine, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Wardah Rabbani
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - David Baker
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Emma S Chambers
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Paul E Pfeffer
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Respiratory Medicine, Barts Health NHS Trust, London, UK
| | - Angray S Kang
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Oral Immunobiology and Regenerative Medicine, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
2
|
Vakrakou AG, Brinia ME, Alexaki A, Koumasopoulos E, Stathopoulos P, Evangelopoulos ME, Stefanis L, Stadelmann-Nessler C, Kilidireas C. Multiple faces of multiple sclerosis in the era of highly efficient treatment modalities: Lymphopenia and switching treatment options challenges daily practice. Int Immunopharmacol 2023; 125:111192. [PMID: 37951198 DOI: 10.1016/j.intimp.2023.111192] [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: 09/03/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/13/2023]
Abstract
The expanded treatment landscape in relapsing-remitting multiple sclerosis (MS) has resulted in highly effective treatment options and complexity in managing disease- or drug-related events during disease progression. Proper decision-making requires thorough knowledge of the immunobiology of MS itself and an understanding of the main principles behind the mechanisms that lead to secondary autoimmunity affecting organs other than the central nervous system as well as opportunistic infections. The immune system is highly adapted to both environmental and disease-modifying agents. Immune reconstitution following cell depletion or cell entrapment therapies eliminates pathogenic aspects of the disease but can also lead to distorted immune responses with harmful effects. Atypical relapses occur with second-line treatments or after their discontinuation and require appropriate clinical decisions. Lymphopenia is a result of the mechanism of action of many drugs used to treat MS. However, persistent lymphopenia and cell-specific lymphopenia could result in disease exacerbation, secondary autoimmunity, or the emergence of opportunistic infections. Clinicians treating patients with MS should be aware of the multiple faces of MS under novel, efficient treatment modalities and understand the intricate brain-immune cell interactions in the context of an altered immune system. MS relapses and disease progression still occur despite the current treatment modalities and are mediated either by failure to control effector mechanisms inherent to MS pathophysiology or by new drug-related mechanisms. The multiple faces of MS due to the highly adapted immune system of patients impose the need for appropriate switching therapies that safeguard disease remission and further clinical improvement.
Collapse
Affiliation(s)
- Aigli G Vakrakou
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Department of Neuropathology, University of Göttingen Medical Center, Göttingen, Germany.
| | - Maria-Evgenia Brinia
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Alexaki
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Koumasopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panos Stathopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Eleftheria Evangelopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Leonidas Stefanis
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Constantinos Kilidireas
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Department of Neurology, Henry Dunant Hospital Center, Athens, Greece
| |
Collapse
|
3
|
Abstract
The development of antidrug antibodies (ADAs) is a major problem in several recombinant protein therapies used in the treatment of multiple sclerosis (MS). The etiology of ADAs is multifaceted. The predisposition for a breakdown of immune tolerance is probably genetically determined, and many factors may contribute to the immunogenicity, including structural properties, formation of aggregates, and presence of contaminants and impurities from the industrial manufacturing process. ADAs may have a neutralizing capacity and can reduce or abrogate the bioactivity and therapeutic efficacy of the drug and cause safety issues. Interferon (IFN)-β was the first drug approved for the treatment of MS, and-although it is generally recognized that neutralizing antibodies (NAbs) appear and potentially have a negative effect on therapeutic efficacy-the use of routine measurements of NAbs and the interpretation of the presence of NAbs has been debated at length. NAbs appear after 9-18 months of therapy in up to 40% of patients treated with IFNβ, and the frequency and titers of NAbs depend on the IFNβ preparation. Although all pivotal clinical trials of approved IFNβ products in MS exhibited a detrimental effect of NAbs after prolonged therapy, some subsequent studies did not observe clinical effects from NAbs, which led to the claim that NAbs did not matter. However, it is now largely agreed that persistently high titers of NAbs indicate an abrogation of the biological response and, hence, an absence of therapeutic efficacy, and this observation should lead to a change of therapy. Low and medium titers are ambiguous, and treatment decisions should be guided by determination of in vivo messenger RNA myxovirus resistance protein A induction after IFNβ administration and clinical disease activity. During treatment with glatiramer acetate, ADAs occur frequently but do not appear to adversely affect treatment efficacy or result in adverse events. ADAs occur in approximately 5% of patients treated with natalizumab within 6 months of therapy, and persistent NAbs are associated with a lack of efficacy and acute infusion-related reactions and should instigate a change of therapy. When using the anti-CD20 monoclonal antibodies ocrelizumab and ofatumumab in the treatment of MS, it is not necessary to test for NAbs as these occur very infrequently. Alemtuzumab is immunogenic, but routine measurements of ADAs are not recommended as the antibodies in the pivotal 2-year trials at the population level did not influence lymphocyte depletion or repopulation, efficacy, or safety. However, in some individuals, NAbs led to poor lymphocyte depletion.
Collapse
|
4
|
Palmeri S, Ponzano M, Ivaldi F, Signori A, Lapucci C, Casella V, Ferrò MT, Vigo T, Inglese M, Mancardi GL, Uccelli A, Laroni A. Impact of Natural Killer (NK) Cells on Immune Reconstitution, and Their Potential as a Biomarker of Disease Activity, in Alemtuzumab-Treated Patients with Relapsing Remitting Multiple Sclerosis: An Observational Study. CNS Drugs 2022; 36:83-96. [PMID: 34894339 DOI: 10.1007/s40263-021-00875-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Defining immune mechanisms leading to multiple sclerosis (MS) is difficult, due to the great inter-individual difference in immune system responses. The anti-CD52 antibody alemtuzumab transiently abolishes differences in immune parameters among individuals, allowing analysis of subsequent immune cell repopulation patterns, and their possible role in MS. OBJECTIVE To evaluate the correlation between innate and adaptive immune cell subsets and disease activity in MS in the context of treatment with alemtuzumab. METHODS A two-center observational cohort of patients treated with alemtuzumab underwent immune profiling of T, B, and natural killer (NK) cells, biomarker, clinical and radiological follow-up. RESULTS After treatment, the percentage of NK and B cells increased; NK, T- and B-cell populations underwent a profound rearrangement. Within the effector T-cell compartment, treatment led to a transient decrease, followed by an increase, of T-helper 1 cells, and to a transient decrease of T-helper 17 cells. Within the T-regulatory compartment, naïve T-regulatory cells increased. Within the B-cell compartment, memory B cells and mature B cells decreased, whereas transitional B cells increased. Within the NK cell compartment, CD56bright NK cells increased. Subjects without disease activity had a greater decrease in serum NfL and greater NK cell/CD3+ T cell ratio. NK cell numbers at baseline and after treatment influenced reconstitution of T and B cells, being inversely correlated with the reconstitution of proinflammatory CD3+ T cells and mature B cells, and directly correlated to the increase in transitional B cells. CONCLUSIONS The results of this study provide novel evidence that NK cells influence reconstitution of adaptive immune cells upon alemtuzumab and that patients with a successful response to alemtuzumab have an early immune reconstitution dominated by NK cells.
Collapse
Affiliation(s)
- Serena Palmeri
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo Daneo 3, 16132, Genoa, Italy.,University of Genova and IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marta Ponzano
- Department of Health Sciences, Section of Biostatistics, University of Genova, Genoa, Italy
| | - Federico Ivaldi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo Daneo 3, 16132, Genoa, Italy
| | - Alessio Signori
- Department of Health Sciences, Section of Biostatistics, University of Genova, Genoa, Italy
| | - Caterina Lapucci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo Daneo 3, 16132, Genoa, Italy.,IRRCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, Genoa, Italy
| | - Valentina Casella
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo Daneo 3, 16132, Genoa, Italy
| | - Maria Teresa Ferrò
- Neuroimmunology, Center for Multiple Sclerosis, Cerebrovascular Department, ASST Crema, Crema, Italy
| | - Tiziana Vigo
- IRRCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, Genoa, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo Daneo 3, 16132, Genoa, Italy.,IRRCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, Genoa, Italy
| | - Giovanni Luigi Mancardi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo Daneo 3, 16132, Genoa, Italy
| | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo Daneo 3, 16132, Genoa, Italy.,IRRCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, Genoa, Italy
| | - Alice Laroni
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo Daneo 3, 16132, Genoa, Italy. .,IRRCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, Genoa, Italy.
| |
Collapse
|
5
|
Ruck T, Barman S, Schulte-Mecklenbeck A, Pfeuffer S, Steffen F, Nelke C, Schroeter CB, Willison A, Heming M, Müntefering T, Melzer N, Krämer J, Lindner M, Riepenhausen M, Gross CC, Klotz L, Bittner S, Muraro PA, Schneider-Hohendorf T, Schwab N, Meyer zu Hörste G, Goebels N, Meuth SG, Wiendl H. OUP accepted manuscript. Brain 2022; 145:1711-1725. [PMID: 35661859 PMCID: PMC9166548 DOI: 10.1093/brain/awac064] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/04/2022] [Accepted: 01/27/2022] [Indexed: 11/12/2022] Open
Abstract
Alemtuzumab is a monoclonal antibody that causes rapid depletion of CD52-expressing immune cells. It has proven to be highly efficacious in active relapsing–remitting multiple sclerosis; however, the high risk of secondary autoimmune disorders has greatly complicated its use. Thus, deeper insight into the pathophysiology of secondary autoimmunity and potential biomarkers is urgently needed. The most critical time points in the decision-making process for alemtuzumab therapy are before or at Month 12, where the ability to identify secondary autoimmunity risk would be instrumental. Therefore, we investigated components of blood and CSF of up to 106 multiple sclerosis patients before and after alemtuzumab treatment focusing on those critical time points. Consistent with previous reports, deep flow cytometric immune-cell profiling (n = 30) demonstrated major effects on adaptive rather than innate immunity, which favoured regulatory immune cell subsets within the repopulation. The longitudinally studied CSF compartment (n = 18) mainly mirrored the immunological effects observed in the periphery. Alemtuzumab-induced changes including increased numbers of naïve CD4+ T cells and B cells as well as a clonal renewal of CD4+ T- and B-cell repertoires were partly reminiscent of haematopoietic stem cell transplantation; in contrast, thymopoiesis was reduced and clonal renewal of T-cell repertoires after alemtuzumab was incomplete. Stratification for secondary autoimmunity did not show clear immununological cellular or proteomic traits or signatures associated with secondary autoimmunity. However, a restricted T-cell repertoire with hyperexpanded T-cell clones at baseline, which persisted and demonstrated further expansion at Month 12 by homeostatic proliferation, identified patients developing secondary autoimmune disorders (n = 7 without secondary autoimmunity versus n = 5 with secondary autoimmunity). Those processes were followed by an expansion of memory B-cell clones irrespective of persistence, which we detected shortly after the diagnosis of secondary autoimmune disease. In conclusion, our data demonstrate that (i) peripheral immunological alterations following alemtuzumab are mirrored by longitudinal changes in the CSF; (ii) incomplete T-cell repertoire renewal and reduced thymopoiesis contribute to a proautoimmune state after alemtuzumab; (iii) proteomics and surface immunological phenotyping do not identify patients at risk for secondary autoimmune disorders; (iv) homeostatic proliferation with disparate dynamics of clonal T- and B-cell expansions are associated with secondary autoimmunity; and (v) hyperexpanded T-cell clones at baseline and Month 12 may be used as a biomarker for the risk of alemtuzumab-induced autoimmunity.
Collapse
Affiliation(s)
- Tobias Ruck
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence to: PD Dr. med. Tobias Ruck Department of Neurology with Institute of Translational Neurology University Hospital Muenster Albert-Schweitzer-Campus 1 D-48149 Muenster, Germany E-mail:
| | - Sumanta Barman
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Andreas Schulte-Mecklenbeck
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Steffen Pfeuffer
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Falk Steffen
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Christopher Nelke
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Christina B. Schroeter
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Alice Willison
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Michael Heming
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Thomas Müntefering
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Nico Melzer
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Julia Krämer
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Maren Lindner
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Marianne Riepenhausen
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Catharina C. Gross
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Paolo A. Muraro
- Department of Brain Sciences, Imperial College London, London, UK
| | - Tilman Schneider-Hohendorf
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Nicholas Schwab
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Gerd Meyer zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
| | - Norbert Goebels
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Sven G. Meuth
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany
- Correspondence may also be addressed to: Univ.-Prof. Prof. h.c. Dr. med. Heinz Wiendl E-mail:
| |
Collapse
|
6
|
Lymphocyte Counts and Multiple Sclerosis Therapeutics: Between Mechanisms of Action and Treatment-Limiting Side Effects. Cells 2021; 10:cells10113177. [PMID: 34831400 PMCID: PMC8625745 DOI: 10.3390/cells10113177] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 01/18/2023] Open
Abstract
Although the detailed pathogenesis of multiple sclerosis (MS) is not completely understood, a broad range of disease-modifying therapies (DMTs) are available. A common side effect of nearly every MS therapeutic agent is lymphopenia, which can be both beneficial and, in some cases, treatment-limiting. A sound knowledge of the underlying mechanism of action of the selected agent is required in order to understand treatment-associated changes in white blood cell counts, as well as monitoring consequences. This review is a comprehensive summary of the currently available DMTs with regard to their effects on lymphocyte count. In the first part, we describe important general information about the role of lymphocytes in the course of MS and the essentials of lymphopenic states. In the second part, we introduce the different DMTs according to their underlying mechanism of action, summarizing recommendations for lymphocyte monitoring and definitions of lymphocyte thresholds for different therapeutic regimens.
Collapse
|
7
|
Marta M, Baker D, Creeke P, Pryce G, Gnanapavan S, Giovannoni G. Antigen-specific tolerization in human autoimmunity: Inhibition of interferon-beta1a anti-drug antibodies in multiple sclerosis: A case report. Mult Scler Relat Disord 2021; 56:103284. [PMID: 34624642 DOI: 10.1016/j.msard.2021.103284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/26/2021] [Accepted: 09/26/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Antigen-specific tolerance in auto-immune diseases is the goal for effective treatment with minimal side-effects. Whilst this is achievable in animal models, notably via intravenous delivery of the model-specific autoantigen following transient CD4 T cell depletion, specific multiple sclerosis autoantigens remain unproven. However, anti-drug antibodies to human therapeutic proteins represent a model human autoimmune condition, which may be used to examine immune-tolerance induction. Some people with MS (PwMS) on interferon-beta1a (IFNβ1a) develop neutralizing antibodies to IFNβ1a that do not disappear in repeated tests over years. METHODS One PwMS was recruited, as part of a planned phase IIa trial (n = 15), who had developed neutralizing antibodies to subcutaneous IFNβ1a. Mitoxantrone (12 mg/m2) was administered as a lymphocyte depleting agent followed by four days of (88 μg/day + three 132 μg/day) intravenous IFNβ1a. Subcutaneous IFNβ1a three times a week was maintained during follow-up. IFNβ1a neutralizing antibody responses in serum were measured during treatment and three-monthly for 12 months. FINDINGS One participant was recruited and, within 6 months of tolerization, the neutralizing antibodies were undetectable. The tolerization treatment was well tolerated. However, the study was terminated after the first enrolment, on ethical grounds, as treatment alternatives became available and the potential risks of mitoxantrone use increased. INTERPRETATION The data suggest that it may be possible to induce antigen-specific tolerance by providing tolerogenic antigen following transient immune depletion. Further studies are warranted. FUNDING The study was supported by an unrestricted research grant from Merck-Serono.
Collapse
Affiliation(s)
- Monica Marta
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom; Clinical Board:Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom.
| | - David Baker
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom.
| | - Paul Creeke
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Gareth Pryce
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Sharmilee Gnanapavan
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom; Clinical Board:Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom.
| | - Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom; Clinical Board:Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom.
| |
Collapse
|
8
|
Baker D, Asardag AN, Quinn OA, Efimov A, Kang AS. Anti-drug antibodies to antibody-based therapeutics in multiple sclerosis. Hum Antibodies 2021; 29:255-262. [PMID: 34397407 DOI: 10.3233/hab-210453] [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] [Indexed: 11/15/2022]
Abstract
Multiple sclerosis is the major demyelinating autoimmune disease of the central nervous system. Relapsing MS can be treated by a number of approved monoclonal antibodies that currently target: CD20, CD25 (withdrawn), CD49d and CD52. These all target potentially pathogenic memory B cell subsets and perhaps functionally inhibit pathogenic T cell function. These consist of chimeric, humanized and fully human antibodies. However, despite humanization it is evident that all of these monoclonal antibodies can induce binding and neutralizing antibodies ranging from < 1% to over 80% within a year of treatment. Importantly, it is evident that monitoring these allow prediction of future treatment-failure in some individuals and treatment cessation and switching therefore potentially limiting disease breakthrough and disability accumulation. In response to the COVID-19 pandemic and the need to avoid hospitals, shortened infusion times and extended dose intervals have been implemented, importantly, subcutaneous delivery of alternative treatments or formulations have been developed to allow for home treatment. Therefore, hospital-based and remote monitoring of ADA could therefore be advantageous to optimize patient responses in the future.
Collapse
Affiliation(s)
- David Baker
- Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - A Nazli Asardag
- Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Olivia A Quinn
- Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alex Efimov
- Camstech Limited, Daresbury Laboratory Science and Technology Facilities Council Sci-Tech, Keckwick, Cheshire, UK
| | - Angray S Kang
- Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Centre for Oral Immunobiology and Regenerative Medicine, Dental Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
9
|
The ocrelizumab phase II extension trial suggests the potential to improve the risk: Benefit balance in multiple sclerosis. Mult Scler Relat Disord 2020; 44:102279. [DOI: 10.1016/j.msard.2020.102279] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 06/01/2020] [Accepted: 06/08/2020] [Indexed: 02/01/2023]
|
10
|
Waddington KE, Papadaki A, Coelewij L, Adriani M, Nytrova P, Kubala Havrdova E, Fogdell-Hahn A, Farrell R, Dönnes P, Pineda-Torra I, Jury EC. Using Serum Metabolomics to Predict Development of Anti-drug Antibodies in Multiple Sclerosis Patients Treated With IFNβ. Front Immunol 2020; 11:1527. [PMID: 32765529 PMCID: PMC7380268 DOI: 10.3389/fimmu.2020.01527] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Neutralizing anti-drug antibodies (ADA) can greatly reduce the efficacy of biopharmaceuticals used to treat patients with multiple sclerosis (MS). However, the biological factors pre-disposing an individual to develop ADA are poorly characterized. Thus, there is an unmet clinical need for biomarkers to predict the development of immunogenicity, and subsequent treatment failure. Up to 35% of MS patients treated with beta interferons (IFNβ) develop ADA. Here we use machine learning to predict immunogenicity against IFNβ utilizing serum metabolomics data. Methods: Serum samples were collected from 89 MS patients as part of the ABIRISK consortium-a multi-center prospective study of ADA development. Metabolites and ADA were quantified prior to and after IFNβ treatment. Thirty patients became ADA positive during the first year of treatment (ADA+). We tested the efficacy of six binary classification models using 10-fold cross validation; k-nearest neighbors, decision tree, random forest, support vector machine and lasso (Least Absolute Shrinkage and Selection Operator) logistic regression with and without interactions. Results: We were able to predict future immunogenicity from baseline metabolomics data. Lasso logistic regression with/without interactions and support vector machines were the most successful at identifying ADA+ or ADA- cases, respectively. Furthermore, patients who become ADA+ had a distinct metabolic response to IFNβ in the first 3 months, with 29 differentially regulated metabolites. Machine learning algorithms could also predict ADA status based on metabolite concentrations at 3 months. Lasso logistic regressions had the greatest proportion of correct classifications [F1 score (accuracy measure) = 0.808, specificity = 0.913]. Finally, we hypothesized that serum lipids could contribute to ADA development by altering immune-cell lipid rafts. This was supported by experimental evidence demonstrating that, prior to IFNβ exposure, lipid raft-associated lipids were differentially expressed between MS patients who became ADA+ or remained ADA-. Conclusion: Serum metabolites are a promising biomarker for prediction of ADA development in MS patients treated with IFNβ, and could provide novel insight into mechanisms of immunogenicity.
Collapse
Affiliation(s)
- Kirsty E. Waddington
- Centre for Rheumatology, University College London, London, United Kingdom
- Centre for Cardiometabolic and Vascular Medicine, University College London, London, United Kingdom
| | - Artemis Papadaki
- Centre for Rheumatology, University College London, London, United Kingdom
| | - Leda Coelewij
- Centre for Rheumatology, University College London, London, United Kingdom
- Centre for Cardiometabolic and Vascular Medicine, University College London, London, United Kingdom
| | - Marsilio Adriani
- Centre for Rheumatology, University College London, London, United Kingdom
| | - Petra Nytrova
- Department of Neurology and Centre of Clinical Neuroscience, General University Hospital and First Faculty of Medicine, Charles University in Prague, Prague, Czechia
| | - Eva Kubala Havrdova
- Department of Neurology and Centre of Clinical Neuroscience, General University Hospital and First Faculty of Medicine, Charles University in Prague, Prague, Czechia
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Center for Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Rachel Farrell
- Department of Neuroinflammation, University College London, Institute of Neurology and National Hospital of Neurology and Neurosurgery, London, United Kingdom
| | - Pierre Dönnes
- Centre for Rheumatology, University College London, London, United Kingdom
- Scicross AB, Skövde, Sweden
| | - Inés Pineda-Torra
- Centre for Cardiometabolic and Vascular Medicine, University College London, London, United Kingdom
| | - Elizabeth C. Jury
- Centre for Rheumatology, University College London, London, United Kingdom
| |
Collapse
|
11
|
Saxena G, Moore JM, Jones M, Pryce G, Ali L, Leisegang GR, Vijay V, Loveless S, Robertson NP, Schmierer K, Giovannoni G, Gnananpavan S, Baker D, Tallantyre EC, Kang AS. Detecting and predicting neutralization of alemtuzumab responses in MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:7/4/e767. [PMID: 32499328 PMCID: PMC7286660 DOI: 10.1212/nxi.0000000000000767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/13/2020] [Indexed: 11/15/2022]
Abstract
Objective To test the hypothesis that antidrug antibodies (ADAs) against alemtuzumab could become relevant after repeated treatments for some individuals, possibly explaining occasional treatment resistance. Methods Recombinant alemtuzumab single-chain variable fragment antibody with a dual tandem nanoluciferase reporter linker was made and used to detect binding ADAs. Alemtuzumab immunoglobulin G Alexa Fluor 488 conjugate was used in a competitive binding cell-based assay to detect neutralizing ADAs. The assays were used to retrospectively screen, blinded, banked serum samples from people with MS (n = 32) who had received 3 or more cycles of alemtuzumab. Lymphocyte depletion was measured between baseline and about 1 month postinfusion. Results The number of individuals showing limited depletion of lymphocytes increased with the number of treatment cycles. Lack of depletion was also a poor prognostic feature for future disease activity. ADA responses were detected in 29/32 (90.6%) individuals. Neutralizing antibodies occurred before the development of limited depletion in 6/7 individuals (18.8% of the whole sample). Preinfusion, ADA levels predicted limited, postinfusion lymphocyte depletion. Conclusions Although ADAs to alemtuzumab have been portrayed as being of no clinical significance, alemtuzumab-specific antibodies appear to be clinically relevant for some individuals, although causation remains to be established. Monitoring of lymphocyte depletion and the antidrug response may be of practical value in patients requiring additional cycles of alemtuzumab. ADA detection may help to inform on retreatment or switching to another treatment.
Collapse
Affiliation(s)
- Gauri Saxena
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - James M Moore
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Meleri Jones
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Gareth Pryce
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Liaqat Ali
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Georgia R Leisegang
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Vivek Vijay
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Samantha Loveless
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Neil P Robertson
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Klaus Schmierer
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Gavin Giovannoni
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Sharmilee Gnananpavan
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - David Baker
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Emma C Tallantyre
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom
| | - Angray S Kang
- From the Blizard Institute (G.S., M.J., G.P., L.A., G.R.L., V.V., K.S., G.G., S.G., D.B., A.S.K.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Division of Psychological Medicine and Clinical Neurosciences (J.M.M., S.L., N.P.R., E.C.T.), Cardiff University School of Medicine, United Kingdom; Department of Biological Sciences (L.A.), National University of Medical Sciences, Rawalpindi, Pakistan; Centre for Oral Immunobiology and Regenerative Medicine (G.R.L., A.S.K.), Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; Clinical Board:Medicine (Neuroscience) (V.V., K.S., G.G., S.G.), The Royal London Hospital, Barts Health NHS Trust; and Welsh Neuroscience Research Tissue Bank (S.L., N.P.R.), Cardiff University, United Kingdom.
| |
Collapse
|
12
|
Rolla S, Maglione A, De Mercanti SF, Clerico M. The Meaning of Immune Reconstitution after Alemtuzumab Therapy in Multiple Sclerosis. Cells 2020; 9:E1396. [PMID: 32503344 PMCID: PMC7348777 DOI: 10.3390/cells9061396] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/02/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
Alemtuzumab is a monoclonal antibody that binds to CD52, a protein present on the surface of mature lymphocytes, but not on the stem cells from which these lymphocytes are derived. It is currently used as an immune reconstitution therapy in patients with relapsing-remitting multiple sclerosis. Alemtuzumab treatment is an intermittent infusion that induces long-term remission of Multiple Sclerosis also in the treatment-free period. After the robust T and B cell depletion induced by alemtuzumab, the immune system undergoes radical changes during its reconstitution. In this review, we will discuss the current knowledge on the reconstitution of the lymphocyte repertoire after alemtuzumab treatment and how it could affect the development of side effects, which led to its temporary suspension by the European Medical Agency.
Collapse
Affiliation(s)
- Simona Rolla
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano (TO), Italy; (A.M.); (S.F.D.M.); (M.C.)
| | | | | | | |
Collapse
|
13
|
CD56bright Natural Killer Cells: A Possible Biomarker of Different Treatments in Multiple Sclerosis. J Clin Med 2020; 9:jcm9051450. [PMID: 32414131 PMCID: PMC7291063 DOI: 10.3390/jcm9051450] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/05/2020] [Accepted: 05/08/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system, which leads, in many cases, to irreversible disability. More than 15 disease-modifying treatments (DMTs) are available for the treatment of MS. Clinical activity or activity at magnetic resonance imaging (MRI) are now used to assess the efficacy of DMTs, but are negative prognostic factors per se. Therefore, a biomarker permitting us to identify patients who respond to treatment before they develop clinical/radiological signs of MS activity would be of high importance. The number of circulating CD56bright natural killer (NK) cells may be such a biomarker. CD56bright NK cells are a regulatory immune population belonging to the innate immune system. The number of CD56bright NK cells increases upon treatment with interferon-beta, alemtuzumab, dimethyl fumarate, after autologous hematopoietic stem cell transplantation, and is higher in those who respond to fingolimod. In some cases, an increased number of CD56bright NK cells is associated with an increase in their regulatory function. In the current review, we will evaluate the known effect on CD56bright NK cells of DMTs for MS, and will discuss their possible role as a biomarker for treatment response in MS.
Collapse
|
14
|
Soleimani B, Murray K, Hunt D. Established and Emerging Immunological Complications of Biological Therapeutics in Multiple Sclerosis. Drug Saf 2020; 42:941-956. [PMID: 30830572 DOI: 10.1007/s40264-019-00799-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Biologic immunotherapies have transformed the treatment landscape of multiple sclerosis. Such therapies include recombinant proteins (interferon beta), as well as monoclonal antibodies (natalizumab, alemtuzumab, daclizumab, rituximab and ocrelizumab). Monoclonal antibodies show particular efficacy in the treatment of the inflammatory phase of multiple sclerosis. However, the immunological perturbations caused by biologic therapies are associated with significant immunological adverse reactions. These include development of neutralising immunogenicity, secondary immunodeficiency and secondary autoimmunity. These complications can affect the balance of risks and benefits of biologic agents, and 2018 saw the withdrawal from the market of daclizumab, an anti-CD25 monoclonal antibody, due to concerns about the development of severe, unpredictable autoimmunity. Here we review established and emerging risks associated with multiple sclerosis biologic agents, with an emphasis on their immunological adverse effects. We also discuss the specific challenges that multiple sclerosis biologics pose to drug safety systems, and the potential for improvements in safety frameworks.
Collapse
Affiliation(s)
| | - Katy Murray
- Anne Rowling Clinic, University of Edinburgh, Edinburgh, UK
| | - David Hunt
- Anne Rowling Clinic, University of Edinburgh, Edinburgh, UK. .,MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
15
|
Ali L, Saxena G, Jones M, Leisegang GR, Gammon L, Gnanapavan S, Giovannoni G, Schmierer K, Baker D, Kang AS. A cell-based assay for the detection of neutralizing antibodies against alemtuzumab. Biotechniques 2020; 68:185-190. [PMID: 32096651 PMCID: PMC7177201 DOI: 10.2144/btn-2019-0122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Aim: The humanized anti-CD52 monoclonal antibody alemtuzumab depletes lymphocytes and is currently used to treat relapsing multiple sclerosis. During treatment, anti-alemtuzumab antibodies may develop and reduce effective lymphocyte depletion in future treatment cycles. Results: Alemtuzumab–Alexa Fluor 488 conjugate binding to the CHO-CD52 cell surface was inhibited by anti-alemtuzumab antibodies. Conclusion: In this proof-of-concept study, a CHO-CD52 cell line has been developed and used to detect the presence of anti-alemtuzumab neutralizing antibodies. This platform provides the basis of an assay for routine screening of serum for neutralizing antibodies from patients treated with alemtuzumab. We developed a competition assay between alemtuzumab and neutralizing antibodies to adherent CD52-expressing cells. The anti-alemtuzumab antibodies inhibit alemtuzumab–Alexa Fluor 488 binding to cell surface CD52. Reduction in the fluorescence signal is proportional the amount of antidrug antibody in the serum sample. Therapeutic monoclonal antibodies are currently used for the treatment of numerous diseases and conditions, including relapsing multiple sclerosis, and are the most advanced targeted therapies available. However, they all have the potential to cause immunogenic reactions and generate antibodies that bind to the drug and reduce its therapeutic efficacy. As a result, patients do not receive the expected benefit from treatment, and the effect is cumulative with repeat dosing. The timely detection of antidrug antibodies has the potential to avoid these major risks. Here we describe a cell-based method for detecting anti-alemtuzumab neutralizing antibodies.
Collapse
Affiliation(s)
- Liaqat Ali
- Barts & The London School of Medicine & Dentistry, Blizard Institute, Queen Mary University of London, Whitechapel, London, UK.,Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi 46000, Pakistan
| | - Gauri Saxena
- Barts & The London School of Medicine & Dentistry, Blizard Institute, Queen Mary University of London, Whitechapel, London, UK
| | - Meleri Jones
- Barts & The London School of Medicine & Dentistry, Blizard Institute, Queen Mary University of London, Whitechapel, London, UK
| | - Georgia R Leisegang
- Centre for Oral Immunobiology & Regenerative Medicine, Institute of Dentistry, Barts & the London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Luke Gammon
- Barts & The London School of Medicine & Dentistry, Blizard Institute, Queen Mary University of London, Whitechapel, London, UK
| | - Sharmilee Gnanapavan
- BartsMS, Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AT, UK.,Clinical Board: Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, E1 1BB, UK
| | - Gavin Giovannoni
- BartsMS, Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AT, UK.,Clinical Board: Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, E1 1BB, UK
| | - Klaus Schmierer
- BartsMS, Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AT, UK.,Clinical Board: Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, E1 1BB, UK
| | - David Baker
- BartsMS, Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Angray S Kang
- Centre for Oral Immunobiology & Regenerative Medicine, Institute of Dentistry, Barts & the London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AT, UK
| |
Collapse
|
16
|
Baker D, Ali L, Saxena G, Pryce G, Jones M, Schmierer K, Giovannoni G, Gnanapavan S, Munger KC, Samkoff L, Goodman A, Kang AS. The Irony of Humanization: Alemtuzumab, the First, But One of the Most Immunogenic, Humanized Monoclonal Antibodies. Front Immunol 2020; 11:124. [PMID: 32117274 PMCID: PMC7034358 DOI: 10.3389/fimmu.2020.00124] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/17/2020] [Indexed: 12/22/2022] Open
Abstract
Alemtuzumab was designed to reduce the immunogenicity of the parent CD52-specific rat immunoglobulin. Although originally marketed for use in cancer (Mabcampath®), alemtuzumab is currently licensed and formulated for the treatment of relapsing multiple sclerosis (Lemtrada®). Perhaps due to its history as the first humanized antibody, the potential of immunogenicity of the molecule has been considered inconsequential, and anti-drug antibodies (ADA) responses were similarly reported as being clinically insignificant. Nonetheless, despite humanization and depletion of peripheral T and B cells, alemtuzumab probably generates the highest frequency of binding and neutralizing ADA of all humanized antibodies currently in clinical use, and they occur rapidly in a large majority of people with MS (pwMS) on alemtuzumab treatment. These ADA appear to be an inherent issue of the biology of the molecule-and more importantly, the target-such that avoidance of immunogenicity-related effects has been facilitated by the dosing schedule used in clinical practice. At the population level this enables the drug to work in most pwMS, but in some individuals, as we show here, antibody neutralization appears to be sufficiently severe to reduce efficacy and allow disease breakthrough. It is therefore imperative that efficacy of lymphocyte depletion and the anti-drug response is monitored in people requiring additional cycles of treatment, notably following disease breakthrough. This may help inform whether to re-treat or to switch to another disease-modifying treatment.
Collapse
Affiliation(s)
- David Baker
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Liaqat Ali
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | - Gauri Saxena
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gareth Pryce
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Meleri Jones
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Klaus Schmierer
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Clinical Board: Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Clinical Board: Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Sharmilee Gnanapavan
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Clinical Board: Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Kathleen C. Munger
- Department of Neurology, University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, NY, United States
| | - Lawrence Samkoff
- Department of Neurology, University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, NY, United States
| | - Andrew Goodman
- Department of Neurology, University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, NY, United States
| | - Angray S. Kang
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
17
|
GloBody Technology: Detecting Anti-Drug Antibody against VH/VL domains. Sci Rep 2020; 10:1860. [PMID: 32024871 PMCID: PMC7002611 DOI: 10.1038/s41598-020-58041-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/26/2019] [Indexed: 12/23/2022] Open
Abstract
The occurrence of anti-drug antibodies following administration of therapeutic monoclonal antibody to patients is a growing problem that is attracting attention from frontline clinicians. Ideally, an initial indicative point of care test would provide guidance to seek testing approved by the regulatory authorities. Here we describe a platform for the detection of IgG anti-drug antibodies that may provide an initial screen for all therapeutic monoclonal antibodies. Synthetic genes encoding Nanoluciferase polypeptides were inserted between the variable heavy and light domain encoding region of known antibody drugs (alemtuzumab and adalimumab) to generate recombinant single chain GloBodies, which retain the drug antibody paratopes and Nanoluciferase activity. In the presence of anti-drug antibodies, the GloBody is bound by specific IgG in the sample. These complexes are captured on immobilised Protein G and the luciferase activity determined. The amount of light generated being indicative of the anti-drug IgG antibody levels in serum. It should be possible to assemble GloBody reagents for all therapeutic monoclonal antibodies and adapt the capture phase to include additional specific isotypes. The assay has the potential to be developed for use with a drop of blood allowing initial pre-screening in a point of care setting.
Collapse
|
18
|
Be aware that the benefits of biological drugs in multiple sclerosis may be offset by their capacity to cause immunological complications. DRUGS & THERAPY PERSPECTIVES 2020. [DOI: 10.1007/s40267-019-00693-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Keller CW, Ruck T, McHugh D, Pfeuffer S, Gross CC, Korsukewitz C, Melzer N, Klotz L, Meuth SG, Münz C, Nimmerjahn F, Wiendl H, Lünemann JD. Impact of FcγR variants on the response to alemtuzumab in multiple sclerosis. Ann Clin Transl Neurol 2019; 6:2586-2594. [PMID: 31682087 PMCID: PMC6917309 DOI: 10.1002/acn3.50935] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/01/2019] [Accepted: 10/09/2019] [Indexed: 12/12/2022] Open
Abstract
Allelic variants of genes encoding for the Fc gamma receptors IIIA and IIA have been associated with the clinical response to cell‐depleting antibodies in lymphoma patients. Here, we tested the hypothesis that FCGR3A and FCGR2A high‐affinity polymorphisms predict clinical outcomes to alemtuzumab therapy in 85 patients with relapsing‐remitting multiple sclerosis. No differences in clinical and MRI‐based efficacy parameters, the development of severe infusion‐associated reactions and secondary autoimmune diseases during a 2 year follow‐up was observed based on FCGR3A or FCGR2A polymorphisms. This study does not support the use of FCGR genetic variants to predict clinical outcomes to alemtuzumab.
Collapse
Affiliation(s)
- Christian W Keller
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Tobias Ruck
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Donal McHugh
- Laboratory of Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Steffen Pfeuffer
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Catharina C Gross
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Catharina Korsukewitz
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Nico Melzer
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Christian Münz
- Laboratory of Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Jan D Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| |
Collapse
|
20
|
Cost of disease modifying therapies for multiple sclerosis: Is front-loading the answer? J Neurol Sci 2019; 404:19-28. [DOI: 10.1016/j.jns.2019.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 01/10/2023]
|
21
|
De Giglio L, Grimaldi AE, Fubelli F, Marinelli F, Pozzilli C. Advances in preventing adverse events during monoclonal antibody management of multiple sclerosis. Expert Rev Neurother 2019; 19:417-429. [PMID: 31094239 DOI: 10.1080/14737175.2019.1610393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Decades of pharmacological research in Multiple Sclerosis (MS) led to the development of therapeutic Monoclonal Antibodies (MAbs) with many different mechanisms of action (MoA), potentially able to improve disability outcome but also determining a more complex management of patients. Areas covered: When clinicians select MS treatments, they should consider adverse events (AEs) on individual basis to minimize patients' risks. Some AEs are common and can be easily handled, but rare complications should also be taken into account. The aim of this review is to summarize existing evidence and provide practical recommendations for the management of therapeutic MAbs in MS. Expert opinion: The introduction of MAbs revolutionized MS treatment with an improvement in effectiveness. Unfortunately, this has been coupled with a more complex array of AEs needing a tighter surveillance strategy. A close interaction between general practitioners, neurologists, and other specialists is the key for a safer use of such effective drugs.
Collapse
Affiliation(s)
- Laura De Giglio
- a MS Center Sant'Andrea Hospital, Sapienza University of Rome , Rome , Italy.,b Department of Human Neuroscience , Sapienza University of Rome , Rome , Italy
| | | | - Federica Fubelli
- a MS Center Sant'Andrea Hospital, Sapienza University of Rome , Rome , Italy
| | | | - Carlo Pozzilli
- a MS Center Sant'Andrea Hospital, Sapienza University of Rome , Rome , Italy.,b Department of Human Neuroscience , Sapienza University of Rome , Rome , Italy
| |
Collapse
|
22
|
Sorensen PS, Sellebjerg F. Pulsed immune reconstitution therapy in multiple sclerosis. Ther Adv Neurol Disord 2019; 12:1756286419836913. [PMID: 30944586 PMCID: PMC6440030 DOI: 10.1177/1756286419836913] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/02/2019] [Indexed: 12/02/2022] Open
Abstract
Whereas drugs used for maintenance/escalation therapy do not maintain their beneficial effect after cessation of therapy, some new highly effective therapies can show prolonged treatment effects after a short treatment course. Such therapies have been named pulsed immune reconstitution therapies or pulsed immunosuppressive therapies, and typical representatives are alemtuzumab and cladribine. Autologous haematopoietic stem cell transplantation could be considered as the strongest immune reconstitution therapy. Both alemtuzumab and cladribine induce depletion of lymphocytes, and a common mechanism of action is preferential depletion of class-switched and unswitched memory B-cells. Whereas CD-19+ B-lymphocytes repopulate within 6 months, CD4+ T-cells repopulate at a slower rate, taking 1–2 years to reach the lower level of normal. In general, the depletion of lymphocytes is more profound and the repletion of T-cells is slower after alemtuzumab than after cladribine treatment. Both drugs have a strong effect on relapses and magnetic resonance imaging (MRI) activity, and reduce disability worsening. The therapeutic effect is maintained beyond the period of active treatment in a large proportion of patients, which is best documented for alemtuzumab. Adverse effects include reactivation of latent infections such as tuberculosis and risk of herpes zoster. The main disadvantage in alemtuzumab-treated patients is the risk of secondary immune-mediated disorders. Pulsed immune reconstitution therapy is an option as initial therapy in relapsing-remitting multiple sclerosis patients with high disease activity and in patients on treatment with another disease-modifying therapy with significant relapse and/or MRI activity.
Collapse
Affiliation(s)
- Per Soelberg Sorensen
- Department of Neurology 2082, Danish Multiple Sclerosis Center, University of Copenhagen, Rigshospitalet, 9, Blegdamsvej, DK-2100 Copenhagen, Denmark
| | - Finn Sellebjerg
- Department of Neurology, Danish Multiple Sclerosis Center, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
23
|
Baker D, Pryce G, Herrod SS, Schmierer K. Potential mechanisms of action related to the efficacy and safety of cladribine. Mult Scler Relat Disord 2019; 30:176-186. [PMID: 30785074 DOI: 10.1016/j.msard.2019.02.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/09/2019] [Accepted: 02/12/2019] [Indexed: 12/31/2022]
Abstract
Oral cladribine is a novel treatment for relapsing multiple sclerosis (MS). This appears to be a semi-selective immune-reconstitution therapy that induces long-term therapy from short treatment cycles. It has a relatively good safety profile that currently does not require extensive monitoring associated with some continuous immunosuppressive and relatively non-selective immune reconstitution therapies. The efficacy and safety of cladribine relates to its particular physicochemical properties, the function of the lymphocyte subsets that are selectively targeted by the drug and the repopulation kinetics of these subsets. As such, there is marked and long-term depletion of memory B cell subsets, which probably relates to the therapeutic efficacy. This is also coupled with a more limited, but likewise long-term, depletion of CD4 T subsets. There is limited depletion of cells of the innate immune system and modest effects on CD8 and probably plasma cells, which provide immediate and durable protection from infection. Targeting of CD4 T regulatory cells, CD8 T suppressor cells and regulatory B cell subsets appears more limited as these populations recover rapidly and so repopulating pathogenic cells re-emerge into a regulatory environment. This appears to lead to re-establishment of immune-tolerance that produces long-term control of MS. Although this hypothesis contains a number of unknown details, it is based on knowledge about the biology of cladribine, basic immunology and the effects of other high-efficacy B and T cell depleting agents that exhibit stereotyped repopulation behaviours. These concepts are relatively simple to interrogate, and can be modified as new knowledge about the durability of disease control and safety with cladribine emerges.
Collapse
Affiliation(s)
- David Baker
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom.
| | - Gareth Pryce
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Samuel S Herrod
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Klaus Schmierer
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Clinical Board:Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London E1 1BB, United Kingdom
| |
Collapse
|
24
|
Regulatory B and T lymphocytes in multiple sclerosis: friends or foes? AUTOIMMUNITY HIGHLIGHTS 2018; 9:9. [PMID: 30415321 PMCID: PMC6230324 DOI: 10.1007/s13317-018-0109-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Current clinical experience with immunomodulatory agents and monoclonal antibodies in principle has established the benefit of depleting lymphocytic populations in relapsing–remitting multiple sclerosis (RRMS). B and T cells may exert multiple pro-inflammatory actions, but also possess regulatory functions making their role in RRMS pathogenesis much more complex. There is no clear correlation of Tregs and Bregs with clinical features of the disease. Herein, we discuss the emerging data on regulatory T and B cell subset distributions in MS and their roles in the pathophysiology of MS and its murine model, experimental autoimmune encephalomyelitis (EAE). In addition, we summarize the immunomodulatory properties of certain MS therapeutic agents through their effect on such regulatory cell subsets and their relevance to clinical outcomes.
Collapse
|