1
|
Wiesinger A, Bigger B, Giugliani R, Lampe C, Scarpa M, Moser T, Kampmann C, Zimmermann G, Lagler FB. Development of a novel tool for individual treatment trials in mucopolysaccharidosis. J Inherit Metab Dis 2025; 48:e12816. [PMID: 39572375 PMCID: PMC11670214 DOI: 10.1002/jimd.12816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 12/28/2024]
Abstract
Mucopolysaccharidosis (MPS) encompasses a group of genetic lysosomal storage disorders, linked to reduced life expectancy and a significant lack of effective treatment options. Immunomodulatory drugs could have the potential to be a relevant medical approach, as the accumulation of undegraded substances initiates an innate immune response, which leads to inflammation and clinical deterioration. However, immunomodulators are not licensed for this indication. Consequently, we aim to provide evidence advocating fast access to innovative individual treatment trials (ITTs) with immunomodulatory drugs and high-quality evaluation of drug effects by implementing a risk-benefit model tailored for MPS. The iterative methodology of our novel decision analysis framework (DAF) involves three key steps: (i) literature review on promising treatment targets and immunomodulators in MPS; (ii) quantitative risk-benefit assessment (RBA) of selected molecules; (iii) assigning phenotypic profiles and quantitative evaluations. The results facilitate a personalized application of the model and are based on published evidence as well as interdisciplinary experts' consensus and patient perspectives. Four promising immunomodulators have been identified: adalimumab, abatacept, anakinra, and cladribine. An improvement in mobility is most likely with adalimumab, while anakinra is anticipated as a treatment of choice for neuronopathic MPS patients. Nevertheless, a comprehensive RBA should always be completed on an individual basis. Our evidence-based DAF tool for ITTs directly addresses the substantial unmet medical need in MPS and characterizes an initial stride toward precision medicine with immunomodulators.
Collapse
Affiliation(s)
- Anna‐Maria Wiesinger
- Institute of Congenital Metabolic DiseasesParacelsus Medical UniversitySalzburgAustria
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
| | - Brian Bigger
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
- Institute for Regeneration and RepairThe University of EdinburghEdinburghUK
| | - Roberto Giugliani
- Department of Genetics, UFRGS, Medical Genetics Service and Biodiscovery LaboratoryHCPA, INAGEMP, DASA, and Casa dos RarosPorto AlegreBrazil
| | - Christina Lampe
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
- Department of Child Neurology, Epileptology and Social Pediatrics, Center of Rare DiseasesUniversity Hospital GiessenMarburgGermany
| | - Maurizio Scarpa
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
- Regional Coordinating Center for Rare DiseasesUniversity Hospital UdineUdineItaly
| | - Tobias Moser
- Department of NeurologyChristian Doppler University Hospital, Paracelsus Medical UniversitySalzburgAustria
| | | | - Georg Zimmermann
- Team Biostatistics and Big Medical DataIDA Lab Salzburg, Paracelsus Medical UniversitySalzburgAustria
- Department of Artificial Intelligence and Human Interfaces, Faculty of Digital and Analytical SciencesParis Lodron UniversitySalzburgAustria
| | - Florian B. Lagler
- Institute of Congenital Metabolic DiseasesParacelsus Medical UniversitySalzburgAustria
- European Reference Network for Hereditary Metabolic DiseasesMetabERNUdineItaly
| |
Collapse
|
2
|
Wiesinger AM, Bigger B, Giugliani R, Lampe C, Scarpa M, Moser T, Kampmann C, Zimmermann G, Lagler FB. An Innovative Tool for Evidence-Based, Personalized Treatment Trials in Mucopolysaccharidosis. Pharmaceutics 2023; 15:1565. [PMID: 37242808 PMCID: PMC10221776 DOI: 10.3390/pharmaceutics15051565] [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: 04/28/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
Mucopolysaccharidosis (MPS) is a group of rare metabolic diseases associated with reduced life expectancy and a substantial unmet medical need. Immunomodulatory drugs could be a relevant treatment approach for MPS patients, although they are not licensed for this population. Therefore, we aim to provide evidence justifying fast access to innovative individual treatment trials (ITTs) with immunomodulators and a high-quality evaluation of drug effects by implementing a risk-benefit model for MPS. The iterative methodology of our developed decision analysis framework (DAF) consists of the following steps: (i) a comprehensive literature analysis on promising treatment targets and immunomodulators for MPS; (ii) a quantitative risk-benefit assessment (RBA) of selected molecules; and (iii) allocation phenotypic profiles and a quantitative assessment. These steps allow for the personalized use of the model and are in accordance with expert and patient representatives. The following four promising immunomodulators were identified: adalimumab, abatacept, anakinra, and cladribine. An improvement in mobility is most likely with adalimumab, while anakinra might be the treatment of choice for patients with neurocognitive involvement. Nevertheless, a RBA should always be completed on an individual basis. Our evidence-based DAF model for ITTs directly addresses the substantial unmet medical need in MPS and characterizes a first approach toward precision medicine with immunomodulatory drugs.
Collapse
Affiliation(s)
- Anna-Maria Wiesinger
- Institute of Congenital Metabolic Diseases, Paracelsus Medical University, 5020 Salzburg, Austria;
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, 33100 Udine, Italy; (B.B.); (C.L.); (M.S.)
| | - Brian Bigger
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, 33100 Udine, Italy; (B.B.); (C.L.); (M.S.)
- Stem Cell and Neurotherapies, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Roberto Giugliani
- Department of Genetics, Medical Genetics Service and Biodiscovery Laboratory, Portal Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Casa dos Raros, Porto Alegre 90610-261, Brazil;
| | - Christina Lampe
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, 33100 Udine, Italy; (B.B.); (C.L.); (M.S.)
- Department of Child Neurology, Epilepetology and Social Pediatrics, Center of Rare Diseases, University Hospital Giessen/Marburg, 35392 Giessen, Germany
| | - Maurizio Scarpa
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, 33100 Udine, Italy; (B.B.); (C.L.); (M.S.)
- Regional Coordinating Center for Rare Diseases, University Hospital Udine, 33100 Udine, Italy
| | - Tobias Moser
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Christoph Kampmann
- Department of Pediatric Cardiology, University Hospital Mainz, 55131 Mainz, Germany;
| | - Georg Zimmermann
- Team Biostatistics and Big Medical Data, IDA Lab Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria;
- Research and Innovation Management, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Florian B. Lagler
- Institute of Congenital Metabolic Diseases, Paracelsus Medical University, 5020 Salzburg, Austria;
- European Reference Network for Hereditary Metabolic Diseases, MetabERN, 33100 Udine, Italy; (B.B.); (C.L.); (M.S.)
| |
Collapse
|
3
|
Muacevic A, Adler JR. Löfgren Syndrome: Clinical Presentation, Clinical Course, and Literature Review. Cureus 2023; 15:e33651. [PMID: 36788849 PMCID: PMC9912994 DOI: 10.7759/cureus.33651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2023] [Indexed: 01/13/2023] Open
Abstract
Löfgren syndrome is an acute presentation of sarcoidosis that comprises fever, bilateral and symmetric hilar lymphadenopathies, polyarthritis, and erythema. We present the case of a 34-year-old male patient who presented with ankle monoarthritis without a history of a traumatic event. Contralateral ankle arthritis associated with erythema nodosum and fever developed one week later. Laboratory tests showed anemia, thrombocytosis, and elevated inflammatory parameters. A chest CT revealed symmetrical mediastinal and hilar adenopathies. A transbronchial biopsy was compatible with granulomatous lymphadenitis, and the diagnosis of Löfgren syndrome was confirmed. Our case report and literature review emphasize the wide web of mimicry of acute sarcoidosis. Secondary forms of acute sarcoidosis are likely to benefit from additional and more complex immunomodulatory therapies. Close monitoring and follow-up should be conducted because it is possible that these patients experience higher rates of recurrence or relapse.
Collapse
|
4
|
Etemadifar M, Mehri A, Sedaghat N, Salari M, Tavassoli Naini P. Multiple sclerosis in sarcoidosis patients: Two case reports. Clin Case Rep 2022; 10:e6332. [PMID: 36172327 PMCID: PMC9468654 DOI: 10.1002/ccr3.6332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/12/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
Two cases of sarcoidosis referred to our clinic with neurological symptoms. They were diagnosed with multiple sclerosis using non-invasive studies. The first patient refused treatment and died of myocardial infarction 6 months after visiting our clinic. The second received interferon-beta and methotrexate with a favorable outcome after 3 years. Since the possible similar presentation of the two conditions could appear indistinct for certain diagnosis, accurate evaluation of symptoms and paraclinical data can provide the best approach to each condition.
Collapse
Affiliation(s)
- Masoud Etemadifar
- Neurosurgery Research DepartmentAlzahra University HospitalIsfahan University of Medical SciencesIsfahanIran
| | - Armin Mehri
- Neurosurgery Research DepartmentAlzahra University HospitalIsfahan University of Medical SciencesIsfahanIran
| | - Nahad Sedaghat
- Neurosurgery Research DepartmentAlzahra University HospitalIsfahan University of Medical SciencesIsfahanIran
| | - Mehri Salari
- Functional Neurosurgery Research CenterShohada Tajrish Comprehensive Neurosurgical Center of ExcellenceShahid Beheshti University of Medical SciencesTehranIran
| | - Parsa Tavassoli Naini
- Neurosurgery Research DepartmentAlzahra University HospitalIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
5
|
Distinguishing CNS neurosarcoidosis from multiple sclerosis and an approach to “overlap” cases. J Neuroimmunol 2022; 369:577904. [DOI: 10.1016/j.jneuroim.2022.577904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 12/17/2022]
|
6
|
Krajnc N, Bsteh G, Berger T, Mares J, Hartung HP. Monoclonal Antibodies in the Treatment of Relapsing Multiple Sclerosis: an Overview with Emphasis on Pregnancy, Vaccination, and Risk Management. Neurotherapeutics 2022; 19:753-773. [PMID: 35378683 PMCID: PMC8978776 DOI: 10.1007/s13311-022-01224-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 01/10/2023] Open
Abstract
Monoclonal antibodies have become a mainstay in the treatment of patients with relapsing multiple sclerosis (RMS) and provide some benefit to patients with primary progressive MS. They are highly precise by specifically targeting molecules displayed on cells involved in distinct immune mechanisms of MS pathophysiology. They not only differ in the target antigen they recognize but also by the mode of action that generates their therapeutic effect. Natalizumab, an [Formula: see text]4[Formula: see text]1 integrin antagonist, works via binding to cell surface receptors, blocking the interaction with their ligands and, in that way, preventing the migration of leukocytes across the blood-brain barrier. On the other hand, the anti-CD52 monoclonal antibody alemtuzumab and the anti-CD20 monoclonal antibodies rituximab, ocrelizumab, ofatumumab, and ublituximab work via eliminating selected pathogenic cell populations. However, potential adverse effects may be serious and can necessitate treatment discontinuation. Most importantly, those are the risk for (opportunistic) infections, but also secondary autoimmune diseases or malignancies. Monoclonal antibodies also carry the risk of infusion/injection-related reactions, primarily in early phases of treatment. By careful patient selection and monitoring during therapy, the occurrence of these potentially serious adverse effects can be minimized. Monoclonal antibodies are characterized by a relatively long pharmacologic half-life and pharmacodynamic effects, which provides advantages such as permitting infrequent dosing, but also creates disadvantages regarding vaccination and family planning. This review presents an overview of currently available monoclonal antibodies for the treatment of RMS, including their mechanism of action, efficacy and safety profile. Furthermore, we provide practical recommendations for risk management, vaccination, and family planning.
Collapse
Affiliation(s)
- Nik Krajnc
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Gabriel Bsteh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Jan Mares
- Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic
| | - Hans-Peter Hartung
- Department of Neurology, Medical University of Vienna, Vienna, Austria.
- Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic.
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
- Brain and Mind Center, University of Sydney, Sydney, Australia.
| |
Collapse
|
7
|
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
|
8
|
Wurm-Kuczera RI, Buentzel J, Koenig JFL, Legler T, Valk JJ, Hasenkamp J, Jung W, Rademacher JG, Korsten P, Wulf GG. Sarcoidosis Following Hematopoietic Stem Cell Transplantation: Clinical Characteristics and HLA Associations. Front Immunol 2021; 12:746996. [PMID: 34691055 PMCID: PMC8529157 DOI: 10.3389/fimmu.2021.746996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose Extrinsic factors and genetic predisposition contribute to the etiology of sarcoidosis, converging in a phenotype of altered immune response associated with multisystemic inflammatory granulomatous tissue infiltration. Immunological reconstitution after hematopoietic stem cell transplantation (HSCT) may represent a unique window for the pathogenesis of the disease. We describe the incidence, clinicopathological features, and HLA associations of sarcoidosis after HSCT in a single-center cohort of patients, together with data from previously published cases. Methods We retrospectively analyzed clinical characteristics and HLA haplotypes from allogeneic (allo) or autologous (auto) HSCT patients from January 2001 through May 2021 at the University Medicine Goettingen (UMG), and data from previously published cases. Results A total number of 19 patients was identified. These included 4 patients from our center (3 allo HSCT and 1 auto HSCT) and 15 patients from the literature review. Thirteen patients had received an allo HSCT, and six patients had received an auto HSCT. Sarcoidosis occurred after a median interval of 20 (after allo HSCT) and 7 (after auto HSCT) months, respectively. The predominant HLA allele associated with sarcoidosis was HLA DRB1*03:01. Sarcoidosis involved the respiratory tract in 15 patients (three unknown, one without pulmonary involvement), and it was associated with graft-versus-host disease in 7 of 13 patients receiving allo HSCT. None of the donors or patients had a history of sarcoidosis before transplantation. Disease manifestations resolved with standard glucocorticoid treatment without long-term sequelae. Conclusion Sarcoidosis may occur at low frequency during reconstitution of the immune system after HSCT. HLA allele associations reflect the associations observed in the general population, particularly with DRB1*03:01. Further insights into the interplay between Tcell reconstitution and the development of sarcoidosis could also provide novel approaches to an improved understanding of the pathogenesis in sarcoidosis.
Collapse
Affiliation(s)
| | - Judith Buentzel
- Department of Hematology and Medical Oncology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Tobias Legler
- Department of Transfusion Medicine, University Medical Center Goettingen, Goettingen, Germany
| | - Jan-Jakob Valk
- Department of Transfusion Medicine, University Medical Center Goettingen, Goettingen, Germany
| | - Justin Hasenkamp
- Department of Hematology and Medical Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Wolfram Jung
- Department of Hematology and Medical Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Jan-Gerd Rademacher
- Department of Nephrology and Rheumatology, University Medical Center Goettingen, Goettingen, Germany
| | - Peter Korsten
- Department of Nephrology and Rheumatology, University Medical Center Goettingen, Goettingen, Germany
| | - Gerald Georg Wulf
- Department of Hematology and Medical Oncology, University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
9
|
Rauma I, Mustonen T, Seppä JM, Ukkonen M, Männikkö M, Verkkoniemi-Ahola A, Kartau M, Saarinen JT, Luostarinen L, Simula S, Ryytty M, Ahmasalo R, Sipilä JOT, Pieninkeroinen I, Tapiola T, Remes AM, Kuusisto H. Safety of alemtuzumab in a nationwide cohort of Finnish multiple sclerosis patients. J Neurol 2021; 269:824-835. [PMID: 34255182 PMCID: PMC8782800 DOI: 10.1007/s00415-021-10664-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/05/2022]
Abstract
Background Alemtuzumab is an effective disease-modifying therapy (DMT) for highly active multiple sclerosis (MS). However, safety concerns limit its use in clinical practice. Objectives To evaluate the safety of alemtuzumab in a nationwide cohort of Finnish MS patients. Methods In this retrospective case series study, we analyzed the data of all but two MS patients who had received alemtuzumab in Finland until 2019. Data were systematically collected from patient files. Results Altogether 121 patients were identified, most of whom had received previous DMTs (82.6%). Median follow-up time after treatment initiation was 30.3 months and exceeded 24 months in 78 patients. Infusion-associated reactions (IARs) were observed in 84.3%, 57.3%, and 57.1% of patients during alemtuzumab courses 1–3, respectively. Serious adverse events (SAEs) were observed in 32.2% of patients, serious IARs in 12.4% of patients, and SAEs other than IARs in 23.1% of patients. Autoimmune adverse events were observed in 30.6% of patients. One patient died of hemophagocytic lymphohistiocytosis, and one patient died of pneumonia. A previously unreported case of thrombotic thrombocytopenic purpura was documented. Conclusions SAEs were more frequent in the present cohort than in previous studies. Even though alemtuzumab is a highly effective therapy for MS, vigorous monitoring with a long enough follow-up time is advised.
Collapse
Affiliation(s)
- Ilkka Rauma
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland. .,Department of Neurology, Tampere University Hospital, Tampere, Finland. .,Department of Neurology, Seinäjoki Central Hospital, Seinäjoki, Finland.
| | - Tiina Mustonen
- Neuro Center, Kuopio University Hospital, Kuopio, Finland
| | | | - Maritta Ukkonen
- Department of Neurology, Tampere University Hospital, Tampere, Finland
| | - Marianne Männikkö
- Department of Neurology, Seinäjoki Central Hospital, Seinäjoki, Finland
| | - Auli Verkkoniemi-Ahola
- Clinical Neurosciences, Neurology, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Marge Kartau
- Clinical Neurosciences, Neurology, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | | | - Liisa Luostarinen
- Department of Neurology, Päijät-Häme Central Hospital, Lahti, Finland
| | - Sakari Simula
- Department of Neurology, Mikkeli Central Hospital, Mikkeli, Finland
| | - Mervi Ryytty
- Medical Research Center, Oulu University Hospital, Oulu, Finland.,Research Unit of Clinical Neuroscience, University of Oulu, Faculty of Medicine, Oulu, Finland
| | - Riitta Ahmasalo
- Department of Neurology, Lapland Central Hospital, Rovaniemi, Finland
| | - Jussi O T Sipilä
- Department of Neurology, North Karelia Central Hospital, Siun Sote, Joensuu, Finland.,Department of Clinical Neurosciences, University of Turku, Turku, Finland
| | | | - Tero Tapiola
- Department of Neurology, South Karelia Central Hospital, Lappeenranta, Finland
| | - Anne M Remes
- Medical Research Center, Oulu University Hospital, Oulu, Finland.,Research Unit of Clinical Neuroscience, University of Oulu, Faculty of Medicine, Oulu, Finland
| | - Hanna Kuusisto
- Department of Neurology, Tampere University Hospital, Tampere, Finland.,Department of Health and Social Management, University of Eastern Finland, Kuopio, Finland.,Department of Neurology, Kanta-Häme Central Hospital, Hämeenlinna, Finland
| |
Collapse
|
10
|
Erlich-Malona N, Cahill J, Chaudhry S, Martin J, Rizvi S. Cardiac sarcoidosis requiring ICD placement and immune thrombocytopenia following alemtuzumab treatment for multiple sclerosis. Mult Scler Relat Disord 2020; 47:102599. [PMID: 33160137 DOI: 10.1016/j.msard.2020.102599] [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: 10/06/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 10/23/2022]
Abstract
Alemtuzumab, an effective disease-modifying therapy for multiple sclerosis, carries a significant risk of secondary autoimmunity. We present a case of cardiac sarcoidosis and immune thrombocytopenia diagnosed in an MS patient two years after completing alemtuzumab treatment. We hypothesize that alemtuzumab-induced changes to the T regulatory cell population may be implicated in the development of sarcoidosis in MS patients.
Collapse
Affiliation(s)
- Natalie Erlich-Malona
- Brown University Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA.
| | - Jonathan Cahill
- Brown University Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Saima Chaudhry
- Brown University Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Janice Martin
- Brown University Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Syed Rizvi
- Brown University Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| |
Collapse
|
11
|
Love B, McCombe JA. Anemia and sarcoidosis following treatment with alemtuzumab. Mult Scler Relat Disord 2020; 46:102526. [PMID: 32979730 DOI: 10.1016/j.msard.2020.102526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/18/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Brandy Love
- Faculty of Medicine, University of Alberta, Canada.
| | - Jennifer A McCombe
- Division of Neurology, Department of Medicine, University of Alberta, Canada.
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW To critically assess the current landscape of disease-modifying agents for multiple sclerosis (MS). Treatment algorithms will be discussed and studies for new agents in late development or recently approved are analyzed in terms of their impact on current treatment strategies. RECENT FINDINGS A real-world study from Wales suggests that early initiation of highly effective therapy may provide more benefit that an escalation approach in relapsing MS. A study from the MSBase dataset found evidence that early treatment with highly effective therapies decreased the risk of developing secondary progressive MS. Ocrelizumab is highly efficacious in relapsing MS and in a group of patients with primary progressive MS. Another CD20 directed mAb, ofatumumab, is in phase 3. A large study examining extended interval dosing of natalizumab in an attempt to decrease the risk of developing progressive multifocal leukoencephalopathy is underway. Cladribine and alemtuzumab may work by immune reconstitution. Siponimod was recently approved by United States Federal Drug Administration for relapsing MS and active secondary progressive MS. Other S1P receptor modulators are being studied in phase 3 trials for relapsing MS. Cladribine received FDA approval as treatment for relapsing and active secondary progressive MS. Autologous hematopoetic stem-cell transplantation may be an option for treatment-refractory MS. SUMMARY Development of disease-modifying agents in MS continues to be successful. Treatment algorithms need to take new developments into account.
Collapse
|
13
|
Bianco A, Mari PV, Larici AR, Lucchini M, Nociti V, Losavio FA, De Fino C, Cicchetti G, Coraci D, Richeldi L, Mirabella M. Alemtuzumab-induced lung injury in multiple sclerosis: Learning from adversity in three patients. Mult Scler Relat Disord 2020; 37:101450. [DOI: 10.1016/j.msard.2019.101450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 10/06/2019] [Accepted: 10/14/2019] [Indexed: 01/07/2023]
|
14
|
Goischke HK. Vitamin D supplementation for the prevention or depletion of side effects of therapy with alemtuzumab in multiple sclerosis. Ther Clin Risk Manag 2019; 15:891-904. [PMID: 31371976 PMCID: PMC6636607 DOI: 10.2147/tcrm.s188941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/16/2019] [Indexed: 12/22/2022] Open
Abstract
PURPOSE OF REVIEW Not only the multiple sclerosis specialist but also the general neurologist and primary care practitioner are increasingly aware of possible adverse events (AEs) by treatment with alemtuzumab (over 47% risk of secondary autoimmune-mediated diseases). Vitamin D supplementation's effect (VDS) to reduce these autoimmune AEs is poorly performed in routine practice. This article seeks to justify why this simple, inexpensive, patient-friendly therapy should be seriously discussed. RECENT FINDINGS Patients who have developed autoimmunity also show a high basal level of IL-21, a cytokine which increases the growth of auto-reactive T-cells. For side effects such as thyroid dysfunction, autoimmune thrombocytopenia, autoimmune hemolytic anemia, autoimmune hepatitis, diabetes mellitus type 1, and alopecia areata/alopecia totalis, VDS may have an impact on the immunological mechanism, in particular lowering levels of IL-17 and IL-21. SUMMARY The potential role of vitamin D in influencing autoimmune diseases is evident. If a life-threatening side-effect can be prevented by high-dose VDS, it is ethical to initiate this add-on therapy despite contradictory results in studies on the effectiveness of VDS.
Collapse
Affiliation(s)
- Hans-Klaus Goischke
- Independent Research, Internal Medicine, Rehabilitation Medicine, Social Medicine, Bad Brückenau, Bavaria, Germany
| |
Collapse
|
15
|
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
|
16
|
|
17
|
Pfeuffer S. Sarcoidosis following alemtuzumab treatment: Autoimmunity mediated by T cells and interferon-γ. Mult Scler 2018; 24:1783-1784. [DOI: 10.1177/1352458518804124] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Steffen Pfeuffer
- Department of Neurology with Institute of Translational Neurology, University of Muenster, Muenster, Germany
| |
Collapse
|