1
|
Jafari P, Hakimian D, Westerhoff M, Cheng J, Cao W, Najibi Kohnehshahri M, Choi WT, Evaristo G, Graham RP, Liao X, Liu X, Pai RK, Salomao MA, Zhao L, Hart J, Micic D, Semrad CE, Alpert L. The Histologic Spectrum of Rituximab-Associated Common Variable Immunodeficiency-Like Enteropathy. Mod Pathol 2025:100770. [PMID: 40222650 DOI: 10.1016/j.modpat.2025.100770] [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: 11/17/2024] [Revised: 03/09/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025]
Abstract
Rituximab (RTX) is a monoclonal anti-CD20 antibody widely used to treat B-cell neoplasms and autoimmune conditions. RTX has recently been linked to an enteropathy characterized by diarrhea, malabsorption, and hypogammaglobulinemia, closely resembling common variable immunodeficiency (CVID) enteropathy. We present the first dedicated histopathologic assessment of RTX-associated CVID-like enteropathy. Study inclusion criteria were the presence of diarrhea, weight loss, or other gastrointestinal (GI) symptoms in the setting of current/prior RTX use and associated hypogammaglobulinemia. Twenty-two patients (15M:7F; mean age at biopsy/resection, 63.4y) across 9 tertiary medical centers met inclusion criteria and had small bowel (N=20) and/or colon (N=17) specimens (biopsies/resections) available for review; 71.4% of specimens dated from ≤5y of last RTX dose. Cases were systematically evaluated by GI pathologists at each institution. Key histologic features in the small bowel included sparse/absent lamina propria plasma cells (N=10, 50%), intraepithelial lymphocytosis (N=12, 60%), villous atrophy (N=11, 55%), increased crypt apoptotic bodies (N=6, 30%), and active inflammation (N=5, 25%). Common features in the colon included sparse/absent plasma cells (N=7, 41.2%), increased crypt apoptotic bodies (N=7, 41.2%), active inflammation (N=5, 29.4%), and intraepithelial lymphocytosis (N=4, 23.5%). Goblet cell loss was appreciated in small bowel and/or colon specimens from 2 patients. Follow-up biopsies (interval, 2m-4y) were available for 7 patients and largely recapitulated the histology of the index specimens, though 1 patient demonstrated improvement in villous blunting and intraepithelial lymphocytosis. In summary, the histologic spectrum of post-RTX CVID-like enteropathy encompasses lamina propria plasma cell depletion, increased crypt apoptotic bodies, small bowel villous atrophy, and goblet cell loss. While the underlying pathophysiology remains uncertain, the clinicopathologic picture may reflect post-RTX B-cell/plasma cell impairment. Although histologic findings may be subtle and variable, pathologists should be aware of this entity and should seek a history of RTX use in patients whose biopsies exhibit these CVID enteropathy-like features.
Collapse
Affiliation(s)
- Pari Jafari
- Department of Pathology, University of Chicago Medicine.
| | - David Hakimian
- Department of Internal Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago Medicine
| | - Maria Westerhoff
- Department of Pathology, Michigan Medicine, University of Michigan
| | - Jerome Cheng
- Department of Pathology, Michigan Medicine, University of Michigan
| | - Wenqing Cao
- Department of Pathology, New York University Langone Health
| | | | - Won-Tak Choi
- Department of Pathology, University of California San Francisco
| | | | - Rondell P Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic Minnesota
| | - Xiaoyan Liao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center
| | - Xiuli Liu
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine
| | - Rish K Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona
| | - Marcela A Salomao
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona
| | - Lei Zhao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School
| | - John Hart
- Department of Pathology, University of Chicago Medicine
| | - Dejan Micic
- Department of Internal Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago Medicine
| | - Carol E Semrad
- Department of Internal Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago Medicine
| | | |
Collapse
|
2
|
Gnanapavan S, Kang A, Baker D, Giovanonni G. Clinical opinions and case studies on understanding and managing hypogammaglobulinaemia in multiple sclerosis: United Kingdom perspective. Mult Scler Relat Disord 2025; 96:106353. [PMID: 40056762 DOI: 10.1016/j.msard.2025.106353] [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: 09/24/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Immunomodulatory disease-modifying therapies (DMTs) in multiple sclerosis (MS) have implications for secondary immunological effects and immunodeficiencies such as hypogammaglobulinaemia and reductions in immunoglobulin levels. Neurologists and treating healthcare professionals should be aware of hypogammaglobulinaemia and be able to manage resulting complications appropriately. METHODS This paper draws on brief clinical case studies in the United Kingdom to illustrate aspects of immunomodulatory DMTs in MS and aims to inform and increase understanding of the role of immunoglobulins in MS, including the presentation of reduced immunoglobulin levels and the interpretation of haematological parameters. RESULTS AND CONCLUSIONS Hypogammaglobulinaemia and prolonged or substantial reductions in immunoglobulin levels carry clinical implications for people living with MS. UK healthcare professionals working in MS can learn from other immunological specialities concerning the management of infection, vaccination, immunisation and treatment with different DMTs. They should be aware of how hypogammaglobulinaemia in MS presents and how to manage this in the context of DMTs, including the interpretation of haematological parameters and safety.
Collapse
Affiliation(s)
- Sharmilee Gnanapavan
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Department of Neurology, Barts Health NHS Trust, London, UK.
| | - Angray Kang
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, 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
| | - Gavin Giovanonni
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Department of Neurology, Barts Health NHS Trust, London, UK
| |
Collapse
|
3
|
Lapucci C, Frau J, Cocco E, Coghe G, Petracca M, Lanzillo R, Brescia Morra V, Nicoletti CG, Landi D, Marfia G, Vercellino M, Cavalla P, Bianco A, Mirabella M, Torri Clerici V, Tomas E, Ferrò MT, Grossi P, Nozzolillo A, Moiola L, Zaffaroni M, Ronzoni M, Pinardi F, Novi G, Cellerino M, Uccelli A, Inglese M. Ocrelizumab in MS patients with persistence of disease activity after alemtuzumab: A multi-center Italian study. Mult Scler 2024; 30:1151-1162. [PMID: 39143825 DOI: 10.1177/13524585241266509] [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: 08/16/2024]
Abstract
BACKGROUND The reason why some multiple sclerosis (MS) patients show disease activity after alemtuzumab (ALM) is still unclear, but ocrelizumab (OCR) could represent an interesting sequential therapeutic approach. OBJECTIVES To investigate safety and efficacy of OCR in MS patients with disease activity after two ALM courses. METHODS Observational retrospective multi-centers Italian cohort study. RESULTS Seventy-two subjects were included. Mean follow-up (FU) was 2.4 (±1) years. Forty-five patients (62.5%) experienced at least one adverse event (AE), with infections accounting for 96.7% of cases. A reduction in total lymphocytes was observed between OCR start and 6 months FU, driven by BCD19+ lymphocytes depletion (p < 0.001). Immunoglobulin M (IgM) levels decreased between OCR start and 6 months FU (p < 0.001). At 2-year FU, relapse, magnetic resonance imaging (MRI) activity and disability worsening-free survival were 92.1%, 90.8%, and 89.2%. The evidence of inflammatory activity between the two ALM courses was associated with higher risk of relapse, MRI activity, and NEDA-3 status loss in relapsing-remitting multiple sclerosis (RRMS; p = 0.02, p = 0.05, p = 0.01, respectively). CONCLUSIONS OCR after two ALM courses seemed to be safe and effective. Early IgM hypogammaglobulinemia occurred in a high proportion of patients. The evidence of inflammatory activity between ALM courses seemed to increase the risk of MS re-activation on OCR treatment.
Collapse
Affiliation(s)
| | - Jessica Frau
- Centro regionale per la diagnosi e la cura della Sclerosi Multipla, ASL Cagliari, Cagliari, Italy
| | - Eleonora Cocco
- Centro regionale per la diagnosi e la cura della Sclerosi Multipla, ASL Cagliari, Cagliari, Italy
- Department of Medical Sciences and Public Health, Università degli studi di Cagliari, Cagliari, Italy
| | - Giancarlo Coghe
- Centro regionale per la diagnosi e la cura della Sclerosi Multipla, ASL Cagliari, Cagliari, Italy
| | - Maria Petracca
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Roberta Lanzillo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Vincenzo Brescia Morra
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | | | - Doriana Landi
- MS Center, Tor Vergata University of Rome, Rome, Italy
| | | | - Marco Vercellino
- Division of Neurology, Department of Neuroscience and Mental Health, City of Health and Science, University Hospital of Torino, Turin, Italy
| | - Paola Cavalla
- Division of Neurology, Department of Neuroscience and Mental Health, City of Health and Science, University Hospital of Torino, Turin, Italy
| | - Assunta Bianco
- Multiple Sclerosis Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Massimiliano Mirabella
- Multiple Sclerosis Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Centro di Ricerca per la Sclerosi Multipla "Anna Paola Batocchi," Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Torri Clerici
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Eugenia Tomas
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Maria Teresa Ferrò
- Neuroimmunology, Neurological Unit, Cerebrovascular Department, Center for Multiple Sclerosis, ASST Crema, Crema, Italy
| | - Paola Grossi
- Neuroimmunology, Neurological Unit, Cerebrovascular Department, Center for Multiple Sclerosis, ASST Crema, Crema, Italy
| | - Agostino Nozzolillo
- Multiple Sclerosis Center, Neurology Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Lucia Moiola
- Multiple Sclerosis Center, Neurology Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Mauro Zaffaroni
- Centro Sclerosi Multipla, Ospedale di Gallarate-ASST della Valle Olona, Gallarate, Italy
| | - Marco Ronzoni
- U.O. Neurologia, ASST Rhodense, Garbagnate Milanese, Italy
| | - Federica Pinardi
- IRCCS Istituto delle scienze neurologiche di Bologna, UOSI Riabilitazione Sclerosi Multipla Bologna, Bologna, Italy
| | - Giovanni Novi
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maria Cellerino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Antonio Uccelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Matilde Inglese
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| |
Collapse
|
4
|
Bonnan M, Courtade H, Debeugny S. Immunoglobulin levels are higher at multiple sclerosis onset - a part of natural history? Mult Scler Relat Disord 2023; 79:105050. [PMID: 37806232 DOI: 10.1016/j.msard.2023.105050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Changes in immunoglobulin (Ig) levels may occur in association with various drugs targeting immunity, including disease-modifying drugs (DMD) and corticosteroids (CS) used to treat multiple sclerosis (MS). However, kinetics of Ig levels during the natural history of MS is poorly described. OBJECTIVE To describe the natural history of the Ig levels in MS. METHODS Monocentric retrospective study examining changes in Ig levels in relation with CS intake in a series of 304 consecutive MS patients (and 1204 samples) followed or hospitalized for 7 years in a single centre. Ig levels are routinely collected in MS patients followed in our centre. RESULTS IgG levels were higher in samples taken at diagnosis than in those taken after the onset of MS symptoms. This effect was also observed in patients remaining free of DMD or CS since onset. On the other hand, overall Ig levels remained stable across fixed time points ranging from 1 to 20 years after onset CONCLUSION: An unanticipated finding of this study was the transient higher IgG levels in samples taken at onset, which suggests that strong inflammatory processes may occur early.
Collapse
Affiliation(s)
- Mickael Bonnan
- Service de neurologie, Hôpital Delafontaine, Saint-Denis 93200, France.
| | - Henri Courtade
- Laboratoire de Biologie Médicale, Centre Hospitalier de Pau, Pau, France
| | - Stéphane Debeugny
- Département d'Information Médicale, Centre Hospitalier de Pau, Pau, France
| |
Collapse
|
5
|
Londoño AC, Mora CA. Continued dysregulation of the B cell lineage promotes multiple sclerosis activity despite disease modifying therapies. F1000Res 2023; 10:1305. [PMID: 37655229 PMCID: PMC10467621 DOI: 10.12688/f1000research.74506.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 09/02/2023] Open
Abstract
A clear understanding of the origin and role of the different subtypes of the B cell lineage involved in the activity or remission of multiple sclerosis (MS) is important for the treatment and follow-up of patients living with this disease. B cells, however, are dynamic and can play an anti-inflammatory or pro-inflammatory role, depending on their milieu. Depletion of B cells has been effective in controlling the progression of MS, but it can have adverse side effects. A better understanding of the role of the B cell subtypes, through the use of surface biomarkers of cellular activity with special attention to the function of memory and other regulatory B cells (Bregs), will be necessary in order to offer specific treatments without inducing undesirable effects.
Collapse
Affiliation(s)
- Ana C. Londoño
- Neurologia y Neuroimagen, Instituto Neurologico de Colombia (INDEC), Medellin, Antioquia, Colombia
| | - Carlos A. Mora
- Spine & Brain Institute, Ascension St. Vincent's Riverside Hospital, Jacksonville, FL, 32204, USA
| |
Collapse
|
6
|
Rolfes L, Pfeuffer S, Skuljec J, He X, Su C, Oezalp SH, Pawlitzki M, Ruck T, Korsen M, Kleinschnitz K, Aslan D, Hagenacker T, Kleinschnitz C, Meuth SG, Pul R. Immune Response to Seasonal Influenza Vaccination in Multiple Sclerosis Patients Receiving Cladribine. Cells 2023; 12:cells12091243. [PMID: 37174643 PMCID: PMC10177067 DOI: 10.3390/cells12091243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/17/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
Cladribine has been approved for the treatment of multiple sclerosis (MS) and its administration results in a long-lasting depletion of lymphocytes. As lymphopenia is known to hamper immune responses to vaccination, we evaluated the immunogenicity of the influenza vaccine in patients undergoing cladribine treatment at different stages vs. controls. The antibody response in 90 cladribine-treated MS patients was prospectively compared with 10 control subjects receiving platform immunotherapy (NCT05019248). Serum samples were collected before and six months after vaccination. Response to vaccination was determined by the hemagglutination-inhibition test. Postvaccination seroprotection rates against influenza A were comparable in cladribine-treated patients and controls (H1N1: 94.4% vs. 100%; H3N2: 92.2% vs. 90.0%). Influenza B response was lower in the cladribine cohort (61.1% vs. 80%). The increase in geometric mean titers was lower in the cladribine group vs. controls (H1N1: +98.5 vs. +188.1; H3N2: +225.3 vs. +300.0; influenza B: +40.0 vs. +78.4); however, titers increased in both groups for all strains. Seroprotection was achieved irrespective of vaccination timing and lymphocyte subset counts at the time of vaccination in the cladribine cohort. To conclude, cladribine-treated MS patients can mount an adequate immune response to influenza independently of treatment duration and time interval to the last cladribine administration.
Collapse
Affiliation(s)
- Leoni Rolfes
- Department of Neurology, HeinrichHeine University Düsseldorf, 40225 Duesseldorf, Germany
| | - Steffen Pfeuffer
- Department of Neurology, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Jelena Skuljec
- Department of Neurology and Center for Translational Neuro and Behavioral Science, University Medicine Essen, 45127 Essen, Germany
- Center for Translational Neuro and Behavioral Sciences (C-TNBS), University Medicine Essen, 45127 Essen, Germany
| | - Xia He
- Department of Neurology and Center for Translational Neuro and Behavioral Science, University Medicine Essen, 45127 Essen, Germany
- Center for Translational Neuro and Behavioral Sciences (C-TNBS), University Medicine Essen, 45127 Essen, Germany
| | - Chuanxin Su
- Department of Neurology and Center for Translational Neuro and Behavioral Science, University Medicine Essen, 45127 Essen, Germany
- Center for Translational Neuro and Behavioral Sciences (C-TNBS), University Medicine Essen, 45127 Essen, Germany
| | - Sinem-Hilal Oezalp
- Department of Neurology and Center for Translational Neuro and Behavioral Science, University Medicine Essen, 45127 Essen, Germany
- Center for Translational Neuro and Behavioral Sciences (C-TNBS), University Medicine Essen, 45127 Essen, Germany
| | - Marc Pawlitzki
- Department of Neurology, HeinrichHeine University Düsseldorf, 40225 Duesseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, HeinrichHeine University Düsseldorf, 40225 Duesseldorf, Germany
| | - Melanie Korsen
- Department of Neurology, HeinrichHeine University Düsseldorf, 40225 Duesseldorf, Germany
| | - Konstanze Kleinschnitz
- Department of Neurology and Center for Translational Neuro and Behavioral Science, University Medicine Essen, 45127 Essen, Germany
- Center for Translational Neuro and Behavioral Sciences (C-TNBS), University Medicine Essen, 45127 Essen, Germany
| | - Derya Aslan
- Department of Neurology and Center for Translational Neuro and Behavioral Science, University Medicine Essen, 45127 Essen, Germany
- Center for Translational Neuro and Behavioral Sciences (C-TNBS), University Medicine Essen, 45127 Essen, Germany
| | - Tim Hagenacker
- Department of Neurology and Center for Translational Neuro and Behavioral Science, University Medicine Essen, 45127 Essen, Germany
- Center for Translational Neuro and Behavioral Sciences (C-TNBS), University Medicine Essen, 45127 Essen, Germany
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational Neuro and Behavioral Science, University Medicine Essen, 45127 Essen, Germany
- Center for Translational Neuro and Behavioral Sciences (C-TNBS), University Medicine Essen, 45127 Essen, Germany
| | - Sven G Meuth
- Department of Neurology, HeinrichHeine University Düsseldorf, 40225 Duesseldorf, Germany
| | - Refik Pul
- Department of Neurology and Center for Translational Neuro and Behavioral Science, University Medicine Essen, 45127 Essen, Germany
- Center for Translational Neuro and Behavioral Sciences (C-TNBS), University Medicine Essen, 45127 Essen, Germany
| |
Collapse
|
7
|
Freeman L, Longbrake EE, Coyle PK, Hendin B, Vollmer T. High-Efficacy Therapies for Treatment-Naïve Individuals with Relapsing-Remitting Multiple Sclerosis. CNS Drugs 2022; 36:1285-1299. [PMID: 36350491 PMCID: PMC9645316 DOI: 10.1007/s40263-022-00965-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/05/2022] [Indexed: 11/11/2022]
Abstract
There are > 18 distinct disease-modifying therapy (DMT) options covering 10 mechanisms of action currently approved by the US Food and Drug Administration for the treatment of relapsing-remitting multiple sclerosis (RRMS). Given the multitude of available treatment options, and recent international consensus guidelines offering differing recommendations, there is broad heterogeneity in how the DMTs are used in clinical practice. Choosing a DMT for newly diagnosed patients with MS is currently a topic of significant debate in MS care. Historically, an escalation approach to DMT was used for newly diagnosed patients with RRMS. However, the evidence for clinical benefits of early treatment with high-efficacy therapies (HETs) in this population is emerging. In this review, we provide an overview of the DMT options and MS treatment strategies, and discuss the clinical benefits of HETs (including ofatumumab, ocrelizumab, natalizumab, alemtuzumab, and cladribine) in the early stages of MS, along with safety concerns associated with these DMTs. By minimizing the accumulation of neurological damage early in the disease course, early treatment with HETs may enhance long-term clinical outcomes over the lifetime of the patient.
Collapse
Affiliation(s)
- Léorah Freeman
- Department of Neurology, Dell Medical School, The University of Texas at Austin, 1601 Trinity St, Austin, TX, 78701, USA.
| | | | - Patricia K Coyle
- Department of Neurology, Stony Brook University Medical Center, Stony Brook, NY, USA
| | - Barry Hendin
- Banner, University Medicine Neurosciences Clinic, Phoenix, AZ, USA
| | - Timothy Vollmer
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
8
|
Mariottini A, Muraro PA, Lünemann JD. Antibody-mediated cell depletion therapies in multiple sclerosis. Front Immunol 2022; 13:953649. [PMID: 36172350 PMCID: PMC9511140 DOI: 10.3389/fimmu.2022.953649] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
Development of disease-modifying therapies including monoclonal antibody (mAb)-based therapeutics for the treatment of multiple sclerosis (MS) has been extremely successful over the past decades. Most of the mAb-based therapies approved for MS deplete immune cell subsets and act through activation of cellular Fc-gamma receptors expressed by cytotoxic lymphocytes and phagocytes, resulting in antibody-dependent cellular cytotoxicity or by initiation of complement-mediated cytotoxicity. The therapeutic goal is to eliminate pathogenic immune cell components and to potentially foster the reconstitution of a new and healthy immune system. Ab-mediated immune cell depletion therapies include the CD52-targeting mAb alemtuzumab, CD20-specific therapeutics, and new Ab-based treatments which are currently being developed and tested in clinical trials. Here, we review recent developments in effector mechanisms and clinical applications of Ab-based cell depletion therapies, compare their immunological and clinical effects with the prototypic immune reconstitution treatment strategy, autologous hematopoietic stem cell transplantation, and discuss their potential to restore immunological tolerance and to achieve durable remission in people with MS.
Collapse
Affiliation(s)
- Alice Mariottini
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
| | - Paolo A. Muraro
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
- *Correspondence: Jan D. Lünemann,
| |
Collapse
|
9
|
Marsh‐Wakefield F, Juillard P, Ashhurst TM, Juillard A, Shinko D, Putri GH, Read MN, McGuire HM, Byrne SN, Hawke S, Grau GE. Peripheral B-cell dysregulation is associated with relapse after long-term quiescence in patients with multiple sclerosis. Immunol Cell Biol 2022; 100:453-467. [PMID: 35416319 PMCID: PMC9322415 DOI: 10.1111/imcb.12552] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/17/2022]
Abstract
B cells play a major role in multiple sclerosis (MS), with many successful therapeutics capable of removing them from circulation. One such therapy, alemtuzumab, is thought to reset the immune system without the need for ongoing therapy in a proportion of patients. The exact cells contributing to disease pathogenesis and quiescence remain to be identified. We utilized mass cytometry to analyze B cells from the blood of patients with relapse-remitting MS (RRMS) before and after alemtuzumab treatment, and during relapse. A complementary RRMS cohort was analyzed by single-cell RNA sequencing. The R package "Spectre" was used to analyze these data, incorporating FlowSOM clustering, sparse partial least squares-discriminant analysis and permutational multivariate analysis of variance. Immunoglobulin (Ig)A+ and IgG1 + B-cell numbers were altered, including higher IgG1 + B cells during relapse. B-cell linker protein (BLNK), CD40 and CD210 expression by B cells was lower in patients with RRMS compared with non-MS controls, with similar results at the transcriptomic level. Finally, alemtuzumab restored BLNK, CD40 and CD210 expression by IgA+ and IgG1 + B cells, which was altered again during relapse. These data suggest that impairment of IgA+ and IgG1 + B cells may contribute to MS pathogenesis, which can be restored by alemtuzumab.
Collapse
Affiliation(s)
- Felix Marsh‐Wakefield
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
- Liver Injury and Cancer ProgramCentenary InstituteSydneyNSWAustralia
- Human Cancer and Viral Immunology LaboratoryThe University of SydneySydneyNSWAustralia
| | - Pierre Juillard
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
| | - Thomas M Ashhurst
- Sydney Cytometry Core Research FacilityThe University of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
| | - Annette Juillard
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
| | - Diana Shinko
- Sydney Cytometry Core Research FacilityThe University of SydneySydneyNSWAustralia
- Ramaciotti Facility for Human Systems BiologyThe University of SydneySydneyNSWAustralia
| | - Givanna H Putri
- School of Computer ScienceThe University of SydneySydneyNSWAustralia
| | - Mark N Read
- School of Computer ScienceThe University of SydneySydneyNSWAustralia
| | - Helen M McGuire
- Ramaciotti Facility for Human Systems BiologyThe University of SydneySydneyNSWAustralia
- Translational Immunology Group, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
| | - Scott N Byrne
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
- Centre for Immunology and Allergy ResearchThe Westmead Institute for Medical ResearchWestmeadNSWAustralia
| | - Simon Hawke
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
- Central West Neurology and NeurosurgeryOrangeNSWAustralia
| | - Georges E Grau
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
| |
Collapse
|
10
|
Rolfes L, Pfeuffer S, Huntemann N, Schmidt M, Su C, Skuljec J, Aslan D, Hackert J, Kleinschnitz K, Hagenacker T, Pawlitzki M, Ruck T, Kleinschnitz C, Meuth SG, Pul R. Immunological consequences of cladribine treatment in multiple sclerosis: A real-world study. Mult Scler Relat Disord 2022; 64:103931. [DOI: 10.1016/j.msard.2022.103931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
|
11
|
Decreased Intrathecal Concentrations of Free Light Chains Kappa in Multiple Sclerosis Patients Taking Very High Effective Disease-Modifying Treatment. Diagnostics (Basel) 2022; 12:diagnostics12030720. [PMID: 35328273 PMCID: PMC8947149 DOI: 10.3390/diagnostics12030720] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/11/2022] [Accepted: 03/13/2022] [Indexed: 11/30/2022] Open
Abstract
Free light chains kappa (FLCκ) in cerebrospinal fluid (CSF) are a part of the intrathecal immune response. This observational study was conducted to investigate the effects of different disease-modifying therapies (DMT) on the humoral intrathecal immune response in the CSF of patients with multiple sclerosis (MS). FLCκ were analyzed in CSF and serum samples from MS patients taking DMT (n = 60) and those in a control cohort of treatment-naïve MS patients (n = 90). DMT was classified as moderately effective (including INFß-1a, INFß-1b, glatiramer acetate, dimethyl fumarate, teriflunomide, triamcinolone); highly effective (including fingolimod, daclizumab) and very highly effective (alemtuzumab, natalizumab, rituximab/ocrelizumab, mitoxantrone). FLCκ were measured using a nephelometric FLCκ kit. Intrathecal FLCκ and IgG concentrations were assessed in relation to the hyperbolic reference range in quotient diagrams. Intrathecal FLCκ concentrations and IgG concentrations were significantly lower in samples from the cohort of MS patients taking very highly effective DMT than in samples from the cohort of MS patients taking highly effective DMT and in the treatment-naïve cohort (FLCκ: p = 0.004, p < 0.0001 respectively/IgG: p = 0.013; p = 0.021). The reduction in FLCκ could contribute to an anti-inflammatory effect in the CNS through this mechanism. There was no difference in the appearance of CSF-specific oligoclonal bands (p = 0.830). Longitudinal analyses are required to confirm these results.
Collapse
|
12
|
Insights into Modern Therapeutic Approaches in Pediatric Acute Leukemias. Cells 2022; 11:cells11010139. [PMID: 35011701 PMCID: PMC8749975 DOI: 10.3390/cells11010139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/07/2021] [Accepted: 12/15/2021] [Indexed: 02/01/2023] Open
Abstract
Pediatric cancers predominantly constitute lymphomas and leukemias. Recently, our knowledge and awareness about genetic diversities, and their consequences in these diseases, have greatly expanded. Modern solutions are focused on mobilizing and impacting a patient’s immune system. Strategies to stimulate the immune system, to prime an antitumor response, are of intense interest. Amid those types of therapies are chimeric antigen receptor T (CAR-T) cells, bispecific antibodies, and antibody–drug conjugates (ADC), which have already been approved in the treatment of acute lymphoblastic leukemia (ALL)/acute myeloid leukemia (AML). In addition, immune checkpoint inhibitors (ICIs), the pattern recognition receptors (PRRs), i.e., NOD-like receptors (NLRs), Toll-like receptors (TLRs), and several kinds of therapy antibodies are well on their way to showing significant benefits for patients with these diseases. This review summarizes the current knowledge of modern methods used in selected pediatric malignancies and presents therapies that may hold promise for the future.
Collapse
|
13
|
Szepanowski F, Warnke C, Meyer Zu Hörste G, Mausberg AK, Hartung HP, Kleinschnitz C, Stettner M. Secondary Immunodeficiency and Risk of Infection Following Immune Therapies in Neurology. CNS Drugs 2021; 35:1173-1188. [PMID: 34657228 PMCID: PMC8520462 DOI: 10.1007/s40263-021-00863-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 12/13/2022]
Abstract
Secondary immunodeficiencies (SIDs) are acquired conditions that may occur as sequelae of immune therapy. In recent years a number of disease-modifying therapies (DMTs) has been approved for multiple sclerosis and related disorders such as neuromyelitis optica spectrum disorders, some of which are frequently also used in- or off-label to treat conditions such as chronic inflammatory demyelinating polyneuropathy (CIDP), myasthenia gravis, myositis, and encephalitis. In this review, we focus on currently available immune therapeutics in neurology to explore their specific modes of action that might contribute to SID, with particular emphasis on their potential to induce secondary antibody deficiency. Considering evidence from clinical trials as well as long-term observational studies related to the patients' immune status and risks of severe infections, we delineate long-term anti-CD20 therapy, with the greatest data availability for rituximab, as a major risk factor for the development of SID, particularly through secondary antibody deficiency. Alemtuzumab and cladribine have relevant effects on circulating B-cell counts; however, evidence for SID mediated by antibody deficiency appears limited and urgently warrants further systematic evaluation. To date, there has been no evidence suggesting that treatment with fingolimod, dimethyl fumarate, or natalizumab leads to antibody deficiency. Risk factors predisposing to development of SID include duration of therapy, increasing age, and pre-existing low immunoglobulin (Ig) levels. Prevention strategies of SID comprise awareness of risk factors, individualized treatment protocols, and vaccination concepts. Immune supplementation employing Ig replacement therapy might reduce morbidity and mortality associated with SIDs in neurological conditions. In light of the broad range of existing and emerging therapies, the potential for SID warrants urgent consideration among neurologists and other healthcare professionals.
Collapse
Affiliation(s)
- Fabian Szepanowski
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Clemens Warnke
- Department of Neurology, University of Cologne, Cologne, Germany
| | | | - Anne K Mausberg
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Medical University Vienna, Vienna, Austria
- Department of Neurology, Palacky University, Olomouc, Czech Republic
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Mark Stettner
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
14
|
Moser T, O’Sullivan C, Puttinger C, Feige J, Pilz G, Haschke-Becher E, Cadamuro J, Oberkofler H, Hitzl W, Harrer A, Kraus J, Trinka E, Wipfler P. Pre-Existing Humoral Immunological Memory Is Retained in Patients with Multiple Sclerosis Receiving Cladribine Therapy. Biomedicines 2021; 9:biomedicines9111584. [PMID: 34829815 PMCID: PMC8615381 DOI: 10.3390/biomedicines9111584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022] Open
Abstract
Cladribine (CLAD) is a lymphodepleting agent approved for active relapsing multiple sclerosis (MS). The impact of CLAD on the adaptive humoral immune system has not sufficiently been studied. This study aimed to assess the influence of CLAD treatment on specific antibody titers to common pathogens. We included 18 MS patients treated with CLAD. Serum IgG antibody levels to measles, mumps, rubella, hepatitis B and varicella zoster virus (VZV), as well as diphtheria and tetanus toxins, were measured prior to the initiation of treatment and at 12 and 24 months after first CLAD administration. Moreover, specimens were longitudinally analyzed regarding absolute blood concentrations of IgG and main lymphocyte subsets. No reduction in antibody levels against measles, mumps, rubella, VZV, hepatitis B, diphtheria toxin and tetanus toxin associated with CLAD treatment was observed. Loss of seroprotection occurred in <1%. We found no significant impact of CLAD on absolute serum IgG levels. Absolute lymphocyte counts were significantly reduced at the end of each treatment year (p < 0.00001 and p < 0.000001). This study suggests that CLAD does not interfere with the pre-existing humoral immunologic memory in terms of pathogen-specific antibody titers.
Collapse
Affiliation(s)
- Tobias Moser
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Center for Cognitive Neuroscience, 5020 Salzburg, Austria (C.P.); (J.F.); (G.P.); (A.H.); (E.T.); (P.W.)
- Correspondence: ; Tel.: +43-57255-30300
| | - Ciara O’Sullivan
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Center for Cognitive Neuroscience, 5020 Salzburg, Austria (C.P.); (J.F.); (G.P.); (A.H.); (E.T.); (P.W.)
| | - Christian Puttinger
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Center for Cognitive Neuroscience, 5020 Salzburg, Austria (C.P.); (J.F.); (G.P.); (A.H.); (E.T.); (P.W.)
| | - Julia Feige
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Center for Cognitive Neuroscience, 5020 Salzburg, Austria (C.P.); (J.F.); (G.P.); (A.H.); (E.T.); (P.W.)
| | - Georg Pilz
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Center for Cognitive Neuroscience, 5020 Salzburg, Austria (C.P.); (J.F.); (G.P.); (A.H.); (E.T.); (P.W.)
| | - Elisabeth Haschke-Becher
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria; (E.H.-B.); (J.C.); (H.O.); (J.K.)
| | - Janne Cadamuro
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria; (E.H.-B.); (J.C.); (H.O.); (J.K.)
| | - Hannes Oberkofler
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria; (E.H.-B.); (J.C.); (H.O.); (J.K.)
| | - Wolfgang Hitzl
- Research Management (RM): Team Biostatistics and Publikation of Clincial Studies, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria;
- Department of Ophthalmology and Optometry Salzburg, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
- Research Program Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Andrea Harrer
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Center for Cognitive Neuroscience, 5020 Salzburg, Austria (C.P.); (J.F.); (G.P.); (A.H.); (E.T.); (P.W.)
- Department of Dermatology and Allergology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Jörg Kraus
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria; (E.H.-B.); (J.C.); (H.O.); (J.K.)
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Eugen Trinka
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Center for Cognitive Neuroscience, 5020 Salzburg, Austria (C.P.); (J.F.); (G.P.); (A.H.); (E.T.); (P.W.)
- Neuroscience Institute, Christian Doppler University Hospital, Paracelsus Medical University and Center for Cognitive Neuroscience, 5020 Salzburg, Austria
| | - Peter Wipfler
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Center for Cognitive Neuroscience, 5020 Salzburg, Austria (C.P.); (J.F.); (G.P.); (A.H.); (E.T.); (P.W.)
| |
Collapse
|
15
|
Correale J, Halfon MJ, Jack D, Rubstein A, Villa A. Acting centrally or peripherally: A renewed interest in the central nervous system penetration of disease-modifying drugs in multiple sclerosis. Mult Scler Relat Disord 2021; 56:103264. [PMID: 34547609 DOI: 10.1016/j.msard.2021.103264] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 09/03/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
With the recent approval of cladribine tablets, siponimod and ozanimod, there has been a renewed interest into the extent to which these current generation disease-modifying therapies (DMTs) are able to cross into the central nervous system (CNS), and how this penetration of the blood-brain barrier (BBB) may influence their ability to treat multiple sclerosis (MS). The integrity of the CNS is maintained by the BBB, blood-cerebrospinal fluid barrier, and the arachnoid barrier, which all play an important role in preserving the immunological environment and homeostasis within the CNS. The integrity of the BBB decreases during the course of MS, with a putative temporal relationship to disease worsening. Furthermore, it is currently considered that progression of the disease is mediated mainly by resident cells of the CNS. The existing literature provides evidence to show that some of the current generation DMTs for MS are able to penetrate the CNS and potentially exert direct effects on CNS-resident cells, in particular the CNS-penetrating prodrugs cladribine and fingolimod, and other sphingosine-1 phosphate receptor modulators; siponimod and ozanimod. Other current generation DMTs appear to be restricted to the periphery due to their high molecular weight or physicochemical properties. As more effective brain penetrant therapies are developed for the treatment of MS, there is a need to understand whether the potential for direct effects within the CNS are of significance, and whether this brings additional benefits over and above treatment effects mediated in the periphery. In turn, this will require an improved understanding of the structure and function of the BBB, the role it plays in MS and subsequent treatments. This narrative review summarizes the data supporting the biological plausibility of a potential benefit from therapeutic molecules entering the CNS, and discusses the potential significance in the current and future treatment of MS.
Collapse
Affiliation(s)
- Jorge Correale
- Department of Neurology, Fleni, Buenos Aires, Argentina.
| | | | - Dominic Jack
- Merck Serono Ltd, Feltham, United Kingdom (an affiliate of Merck KGaA)
| | - Adrián Rubstein
- Merck S.A., Buenos Aires, Argentina (an affiliate of Merck KGaA)
| | - Andrés Villa
- Hospital Ramos Mejía, Universidad de Buenos Aires, Argentina
| |
Collapse
|
16
|
Pawlitzki M, Meuth SG. Immunmodulatorische Therapien bei Multipler Sklerose in der Pandemie. INFO NEUROLOGIE + PSYCHIATRIE 2021. [PMCID: PMC8373422 DOI: 10.1007/s15005-021-2009-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Marc Pawlitzki
- des UniversitätsklinikumMünster, Institut für translationale Neurologie, Albert-Schweitzer-Campus1, 48149 Münster, Germany
| | - Sven G. Meuth
- Klinik für Neurologie, Universitätsklinikum Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| |
Collapse
|
17
|
Jakimovski D, Eckert SP, Zivadinov R, Weinstock-Guttman B. Considering patient age when treating multiple sclerosis across the adult lifespan. Expert Rev Neurother 2021; 21:353-364. [PMID: 33595379 DOI: 10.1080/14737175.2021.1886082] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: The successful development of anti-inflammatory disease-modifying treatments (DMT) significantly improved disease outcomes and longevity of persons with multiple sclerosis (pwMS). However, the shift toward an elderly MS population has resulted with new concerns regarding DMT efficacy and safety.Areas covered: This review summarizes the evidence of an age-based decrease in the efficacy of MS DMTs and increase in pharmacovigilance concerns. The age effects on pathophysiological MS processes, immunosenescence and its relevance to DMT selection or discontinuation are also reviewed. Lastly, the authors discuss the influence of age-associated comorbidities on DMT initiation and drug-induced events.Expert opinion: There is an age discrepancy between pwMS included in regulatory drug trials and an aging real-world MS population. Most trials demonstrate significantly diminished anti-inflammatory efficacy in patients older than 40 years old. Older age is associated with a greater risk for adverse events including serious infections. Age-associated comorbidities influence the risk-benefit analysis and sometimes cause patients to discontinue DMTs. Instead of chronological age cutoffs, therefore, studies should aim at promoting biologically-based age biomarkers.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.,Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| | - Svetlana P Eckert
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.,Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA.,Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Bianca Weinstock-Guttman
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
18
|
Abstract
Alemtuzumab (Lemtrada®) is an anti-CD52 monoclonal antibody approved in the EU for the treatment of highly active relapsing-remitting multiple sclerosis (RRMS). In phase 3 trials in patients with active RRMS, intravenous alemtuzumab was more effective than subcutaneous interferon β-1a in terms of decreasing relapse rates (in treatment-naïve or -experienced patients) and disability progression (treatment-experienced patients). Treatment benefits were maintained over up to 9 years of follow-up, with ≈ 50% of patients not requiring retreatment. The efficacy of alemtuzumab in patients with highly active disease was generally similar to that in the overall population. Alemtuzumab has an acceptable tolerability profile, with infusion-associated reactions, infections and autoimmunity being the main safety and tolerability issues. Current evidence indicates that alemtuzumab is an effective treatment option for adults with highly active RRMS, with an acceptable safety and tolerability profile and convenient treatment regimen.
Collapse
Affiliation(s)
- Yahiya Y Syed
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand.
| |
Collapse
|
19
|
McMasters M, Blair BM, Lazarus HM, Alonso CD. Casting a wider protective net: Anti-infective vaccine strategies for patients with hematologic malignancy and blood and marrow transplantation. Blood Rev 2020; 47:100779. [PMID: 33223246 DOI: 10.1016/j.blre.2020.100779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 10/29/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023]
Abstract
Patients who have hematologic malignancies are at high risk for infections but vaccinations may be effective prophylaxis. The increased infection risk derives from immune defects secondary to malignancy, the classic example being CLL, and chemotherapies and immunotherapy used to treat the malignancies. Therapy of hematologic malignancies is being revolutionized by introduction of novel targeted agents and immunomodulatory medications, improving the survival of patients. At the same time those agents uniquely change the infection risk and response to immunizations. This review will summarize current vaccine recommendations for patients with hematologic malignancies including patients who undergo hematopoietic cell transplant.
Collapse
Affiliation(s)
- Malgorzata McMasters
- Division of Hematologic Malignancy and Bone Marrow Transplant, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215, USA; Harvard Medical School, Boston, MA, USA
| | - Barbra M Blair
- Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Beth Israel Deaconess Medical Center, 110 Francis Street, Suite GB, Boston, MA 02215, USA
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Carolyn D Alonso
- Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Beth Israel Deaconess Medical Center, 110 Francis Street, Suite GB, Boston, MA 02215, USA.
| |
Collapse
|
20
|
Peter HH, Ochs HD, Cunningham-Rundles C, Vinh DC, Kiessling P, Greve B, Jolles S. Targeting FcRn for immunomodulation: Benefits, risks, and practical considerations. J Allergy Clin Immunol 2020; 146:479-491.e5. [PMID: 32896308 PMCID: PMC7471860 DOI: 10.1016/j.jaci.2020.07.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/08/2023]
Abstract
The neonatal fragment crystallizable (Fc) receptor (FcRn) functions as a recycling mechanism to prevent degradation and extend the half-life of IgG and albumin in the circulation. Several FcRn inhibitors selectively targeting IgG recycling are now moving rapidly toward clinical practice in neurology and hematology. These molecules accelerate the destruction of IgG, reducing pathogenic IgG and IgG immune complexes, with no anticipated effects on IgA, IgM, IgE, complement, plasma cells, B cells, or other cells of the innate or adaptive immune systems. FcRn inhibitors have potential for future use in a much wider variety of antibody-mediated autoimmune diseases. Given the imminent clinical use, potential for broader utility, and novel mechanism of action of FcRn inhibitors, here we review data from 4 main sources: (a) currently available activity, safety, and mechanism-of-action data from clinical trials of FcRn inhibitors; (b) other procedures and treatments that also remove IgG (plasma donation, plasma exchange, immunoadsorption); (c) diseases resulting in loss of IgG; and (d) primary immunodeficiencies with potential mechanistic similarities to those induced by FcRn inhibitors. These data have been evaluated to provide practical considerations for the assessment, monitoring, and reduction of any potential infection risk associated with FcRn inhibition, in addition to highlighting areas for future research.
Collapse
Affiliation(s)
- Hans-Hartmut Peter
- Freiburg University Hospital, Centre for Chronic Immunodeficiency, Freiburg, Germany
| | - Hans D Ochs
- Seattle Children's Research Institute, Seattle, Wash; Department of Pediatrics, University of Washington, Seattle, Wash
| | | | - Donald C Vinh
- Division of Infectious Diseases, Department of Medicine and Department of Medical Microbiology, McGill University Health Centre, Montreal, Quebec, Canada; Infectious Diseases & Immunity in Global Health Program, Research Institute-McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, United Kingdom.
| |
Collapse
|
21
|
Korsukewitz C, Reddel SW, Bar-Or A, Wiendl H. Neurological immunotherapy in the era of COVID-19 - looking for consensus in the literature. Nat Rev Neurol 2020; 16:493-505. [PMID: 32641860 PMCID: PMC7341707 DOI: 10.1038/s41582-020-0385-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2020] [Indexed: 01/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is concerning for patients with neuroimmunological diseases who are receiving immunotherapy. Uncertainty remains about whether immunotherapies increase the risk of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or increase the risk of severe disease and death upon infection. National and international societies have developed guidelines and statements, but consensus does not exist in several areas. In this Review, we attempt to clarify where consensus exists and where uncertainty remains to inform management approaches based on the first principles of neuroimmunology. We identified key questions that have been addressed in the literature and collated the recommendations to generate a consensus calculation in a Delphi-like approach to summarize the information. We summarize the international recommendations, discuss them in light of the first available data from patients with COVID-19 receiving immunotherapy and provide an overview of management approaches in the COVID-19 era. We stress the principles of medicine in general and neuroimmunology in particular because, although the risk of viral infection has become more relevant, most of the considerations apply to the general management of neurological immunotherapy. We also give special consideration to immunosuppressive treatment and cell-depleting therapies that might increase susceptibility to SARS-CoV-2 infection but reduce the risk of severe COVID-19.
Collapse
Affiliation(s)
- Catharina Korsukewitz
- Department of Neurology with Institute of Translational Neurology, University of Muenster, Muenster, Germany
| | - Stephen W Reddel
- Department of Neurology, Concord Hospital and The Brain and Mind Centre, University of Sydney, Sydney, Australia
| | - Amit Bar-Or
- Center for Neuroinflammation and Neurotherapeutics and the Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Muenster, Muenster, Germany.
| |
Collapse
|
22
|
Differential Effects of MS Therapeutics on B Cells-Implications for Their Use and Failure in AQP4-Positive NMOSD Patients. Int J Mol Sci 2020; 21:ijms21145021. [PMID: 32708663 PMCID: PMC7404039 DOI: 10.3390/ijms21145021] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/25/2022] Open
Abstract
B cells are considered major contributors to multiple sclerosis (MS) pathophysiology. While lately approved disease-modifying drugs like ocrelizumab deplete B cells directly, most MS medications were not primarily designed to target B cells. Here, we review the current understanding how approved MS medications affect peripheral B lymphocytes in humans. These highly contrasting effects are of substantial importance when considering these drugs as therapy for neuromyelitis optica spectrum disorders (NMOSD), a frequent differential diagnosis to MS, which is considered being a primarily B cell- and antibody-driven diseases. Data indicates that MS medications, which deplete B cells or induce an anti-inflammatory phenotype of the remaining ones, were effective and safe in aquaporin-4 antibody positive NMOSD. In contrast, drugs such as natalizumab and interferon-β, which lead to activation and accumulation of B cells in the peripheral blood, lack efficacy or even induce catastrophic disease activity in NMOSD. Hence, we conclude that the differential effect of MS drugs on B cells is one potential parameter determining the therapeutic efficacy or failure in antibody-dependent diseases like seropositive NMOSD.
Collapse
|
23
|
Otto F, Moser T, Feige J, Seiberl M, Bieler L, Sellner J. Successful disease control with alemtuzumab in MOG-IgG-associated demyelinating disease with MS-phenotype. Mult Scler Relat Disord 2020; 42:102108. [PMID: 32339987 DOI: 10.1016/j.msard.2020.102108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 01/25/2023]
Abstract
Immunoglobulin G (IgG) antibodies against myelin oligodendrocyte glycoprotein in serum denote an emerging autoimmune disorder of the central nervous system. Treatment trials have not been performed so far and anecdotal reports suggest that immunotherapies approved for multiple sclerosis (MS) may not be effective. We report favorable disease control with alemtuzumab, a CD52 depleting antibody approved for active MS, in a 34-year-old woman with the rarer condition of MOG-IgG disease with MS-phenotype. MOG-IgG in serum persisted over the entire observation period of almost five years. This case emphasizes that treatment responses may be distinct for different phenotypes of MOG-IgG associated disease.
Collapse
Affiliation(s)
- Ferdinand Otto
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020.
| | - Tobias Moser
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020.
| | - Julia Feige
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020.
| | - Michael Seiberl
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020
| | - Lara Bieler
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020
| | - Johann Sellner
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, 5020; Department of Neurology, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany; Department of Neurology, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria.
| |
Collapse
|
24
|
Risiken und Chancen von Immuntherapien in Zeiten der Coronavirus-2019-Pandemie. DGNEUROLOGIE 2020. [PMCID: PMC7284681 DOI: 10.1007/s42451-020-00205-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Immuntherapien stellen die essenzielle Grundlage der Behandlung von neuroinflammatorischen Erkrankungen dar. In Zeiten der Coronavirus-2019 (COVID-19)-Pandemie ergibt sich im klinischen Alltag jedoch zunehmend die Frage, ob eine Immuntherapie bei neurologischen Patienten aufgrund des potenziellen Infektionsrisikos eingeleitet, intensiviert, pausiert oder gar beendet werden sollte. Unsicherheit besteht v. a. deshalb, weil verschiedene nationale und internationale Fachgesellschaften diesbezüglich unterschiedliche Empfehlungen veröffentlichten. In diesem Artikel soll ein Überblick über die Wirkmechanismen von Immuntherapien und den daraus abzuleitenden Infektionsrisiken in Bezug auf COVID-19 (durch den Coronavirus verursachte Erkrankung) gegeben werden. Potenzielle Chancen und vorteilhafte Effekte einzelner Substrate in der Akuttherapie von COVID-19 werden diskutiert.
Collapse
|
25
|
Pawlitzki M, Zettl UK, Ruck T, Rolfes L, Hartung HP, Meuth SG. Merits and culprits of immunotherapies for neurological diseases in times of COVID-19. EBioMedicine 2020; 56:102822. [PMID: 32535547 PMCID: PMC7286830 DOI: 10.1016/j.ebiom.2020.102822] [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: 05/01/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
Immunosuppression and immunomodulation are valuable therapeutic approaches for managing neuroimmunological diseases. In times of the Coronavirus disease 2019 (COVID-19) pandemic, clinicians must deal with the question of whether immunotherapy should currently be initiated or discontinued in neurological patients. Uncertainty exists especially because different national medical associations publish different recommendations on the extent to which immunotherapies must be continued, monitored, or possibly switched during the current pandemic. Based on the most recently available data both about the novel coronavirus and the approved immunotherapies for neurological diseases, we provide an updated overview that includes current treatment strategies and the associated COVID-19 risk, but also the potential of immunotherapies to treat COVID-19.
Collapse
Affiliation(s)
- Marc Pawlitzki
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.
| | - Uwe K Zettl
- Department of Neurology, Neuroimmunological Section, University of Rostock, Rostock, Germany
| | - Tobias Ruck
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Leoni Rolfes
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
26
|
Dalmau J. How DIRS is refining concepts. NEUROLOGY - NEUROIMMUNOLOGY NEUROINFLAMMATION 2020; 7:7/2/e677. [PMID: 32033982 PMCID: PMC7051200 DOI: 10.1212/nxi.0000000000000677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Josep Dalmau
- From ICREA-IDIBAPS Hospital Clínic, University of Barcelona, Spain
| |
Collapse
|