1
|
Stino AM, Bumma N, Smith R, Davalos L, Allen J, Ye JC, Pianko M, Campagnaro E, Fierro C, Awad A, Murdock B, Pietrzak M, Loszanski G, Kline DM, Efebera Y, Elsheikh B. Lenalidomide in the treatment of anti-myelin-associated glycoprotein neuropathy: A phase 1 study to identify the maximum tolerated dose. Eur J Neurol 2024; 31:e16164. [PMID: 38015467 PMCID: PMC11235859 DOI: 10.1111/ene.16164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/12/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Anti-myelin-associated glycoprotein (MAG) neuropathy is a debilitating demyelinating polyneuropathy with no approved therapies. Our primary objective was to ascertain lenalidomide safety and maximum tolerated dose (MTD) in anti-MAG neuropathy. METHODS This phase 1b, open-label, single-arm, dose-finding trial was conducted from 2019 through 2022. The original design included a dose-escalation/extension phase followed by a dose-expansion phase. Three doses of lenalidomide were evaluated: 10, 15, and 25 mg. The main outcome was the MTD. RESULTS Eleven patients enrolled (10 men), with a mean age of 67.6 years (SD = 6.18, range 58-77 years) and mean disease duration of 8.5 years (SD = 10.9, range 1-40 years). The study terminated early due to higher-than-expected non-dose-limiting toxicity venous thromboembolism (VTE) events. The calculated MTD was 25 mg (posterior mean of toxicity probability was 0.01 with a 95% credible interval of 0.00, 0.06), but a recommended phase 2 dose of 15 mg was advised. For secondary exploratory outcomes, only EQ-5D (-0.95, 95% CI -1.81 to -0.09) and total IgM (-162 mg/dL, 95% CI -298 to -26) showed signs of improvement by month 12. CONCLUSIONS Lenalidomide was associated with higher-than-expected VTE events in anti-MAG neuropathy patients, despite a calculated MTD of 25 mg. A recommended phase 2 dose of 15 mg was advised. Lenalidomide did not improve disability or impairment at 12 months, although this study was not powered for efficacy. The risks of long term lenalidomide may outweigh benefit for patients with anti-MAG neuropathy. Any future efficacy study should address VTE risk, as current myeloma guidelines appear inadequate. TRIAL REGISTRATION Lenalidomide in Anti-MAG Neuropathy: Phase 1b Study, ClinicalTrials.gov Identifier: NCT03701711, https://clinicaltrials.gov/ct2/show/NCT03701711. First submitted October 10, 2018. First patient enrolled in January 2019.
Collapse
Affiliation(s)
- Amro M. Stino
- Department of Neurology, Division of Neuromuscular MedicineThe University of Michigan Health SystemAnn ArborMichiganUSA
| | - Naresh Bumma
- Department of Internal Medicine, Division of Hematology‐OncologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Rachel Smith
- Center for BiostatisticsThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Long Davalos
- Department of Neurology, Division of Neuromuscular MedicineThe University of Cincinnati Medical CenterCincinnatiOhioUSA
| | - Jeff Allen
- Department of Neurology, Division of Neuromuscular MedicineUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Jing Christine Ye
- Department of Internal Medicine, Division of Hematology‐OncologyThe University of Michigan Health SystemAnn ArborMichiganUSA
| | - Matthew Pianko
- Department of Internal Medicine, Division of Hematology‐OncologyThe University of Michigan Health SystemAnn ArborMichiganUSA
| | - Erica Campagnaro
- Department of Internal Medicine, Division of Hematology‐OncologyThe University of Michigan Health SystemAnn ArborMichiganUSA
| | - Cassandra Fierro
- Department of Neurology, Division of Neuromuscular MedicineThe University of Michigan Health SystemAnn ArborMichiganUSA
| | - Abdelrahman Awad
- Department of Neurology, Division of Neuromuscular MedicineThe University of Michigan Health SystemAnn ArborMichiganUSA
| | - Ben Murdock
- Department of Neurology, Division of Neuromuscular MedicineThe University of Michigan Health SystemAnn ArborMichiganUSA
| | - Maciej Pietrzak
- Department of Biomedical InformaticsThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Gerard Loszanski
- Department of PathologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - David M. Kline
- Department of Biostatistics and Data Science, Division of Public Health SciencesWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | | | - Bakri Elsheikh
- Department of Neurology, Division of Neuromuscular MedicineThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|
2
|
Stino AM, Elsheikh B, Allen JA. Anti-myelin-associated glycoprotein neuropathy: Where do we stand? Muscle Nerve 2023; 68:823-832. [PMID: 37602932 DOI: 10.1002/mus.27954] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 08/22/2023]
Abstract
Myelin-associated glycoprotein (MAG) is a transmembrane glycoprotein concentrated in periaxonal Schwann cell and oligodendroglial membranes of myelin sheaths that serves as an antigen for immunoglobulin M (IgM) monoclonal antibodies. Individuals who harbor anti-MAG antibodies classically develop a progressive autoimmune peripheral neuropathy characterized clinically by ataxia, distal sensory loss, and gait instability, and electrophysiologically by distally accentuated conduction velocity slowing. Although off-label immunotherapy is common, there are currently no proven effective disease-modifying therapeutics, and most patients experience slow accumulation of disability over years and decades. The typically slowly progressive nature of this neuropathy presents unique challenges when trying to find effective anti-MAG therapeutic agents. Drug development has also been hampered by the lack of validated outcome measures that can detect clinically meaningful changes in a reasonable amount of time as well as by the lack of disease activity biomarkers. In this invited review, we provide an update on the state of clinicometric outcome measures and disease activity biomarkers in anti-MAG neuropathy. We highlight the insensitivity of widely used existing clinicometric outcome measures such as the Inflammatory Neuropathy Cause and Treatment (INCAT) disability score as well as the INCAT sensory subscore in anti-MAG neuropathy, referencing the two previous negative randomized controlled clinical trials evaluating rituximab. We then discuss newly emerging candidate therapeutic agents, including tyrosine kinase inhibitors and enhanced B-cell-depleting agents, among others. We conclude with a practical approach to the evaluation and management of anti-MAG neuropathy patients.
Collapse
Affiliation(s)
- Amro Maher Stino
- Department of Neurology, Division of Neuromuscular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Bakri Elsheikh
- Department of Neurology, Division of Neuromuscular Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Jeffrey A Allen
- Department of Neurology, Division of Neuromuscular Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Bozovic I, Perovic V, Basta I, Peric S, Stevic Z, Popadic D, Vukovic I, Stojanov A, Milosevic E. Cytokine Gene Polymorphisms in Patients with Chronic Inflammatory Demyelinating Polyneuropathy. Cells 2023; 12:2033. [PMID: 37626843 PMCID: PMC10453148 DOI: 10.3390/cells12162033] [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: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Innate and adaptive immune responses exert their role in CIDP pathogenesis through cytokine production. Single-nucleotide polymorphisms (SNPs) may alter cytokine gene expression, with a potential influence on the pathogenesis of autoimmune diseases. However, cytokine gene SNPs have not been assessed in CIDP patients yet. We assessed functional SNPs in the genes encoding IL-10 (rs1800896, rs1800871, rs1800872 and rs3024505), IL-6 (rs1800795), TNF (rs1800629 and rs361525), IL-12B (rs3212227), IFN-γ (rs2430561), GM-CSF (rs25882) and IL-17F (rs11465553) in a cohort of 88 CIDP patients and 486 healthy controls (HCs) via qPCR. We found an association of SNP in the IL10 promotor and CIDP occurrence. Major homozygotes (AA) were more frequent in the HCs compared to CIDP patients (p = 0.049), but the GA genotype prevailed among the patients (p = 0.032). A lower frequency of the C allele was observed for rs1800871 and rs1800872 in CIDP patients compared to the HCs (p = 0.048). A higher proportion of A carriers at position -1082 (rs1800896) (presumed to be a low IL-10 producer) was noted in patients with milder disability (low INCAT). All mild-INCAT patients were C carriers for rs1800871 and rs1800872 in IL10 (p = 0.038). Furthermore, the IL6 rs1800795 GG genotype was more frequent in patients (p = 0.049) and the CG heterozygote in the HCs (p = 0.013). Among the CIDP patients, being a G carrier for this SNP was associated with a higher frequency of type 2 diabetes (T2D) compared to being a non-carrier (p = 0.032). Our data indicate a possible association of the IL10 and IL6 SNPs with CIDP, but also with disease severity and T2D occurrence. Given the paucity of CIDP patients, multicentric studies are necessary to draw definite conclusions on these associations.
Collapse
Affiliation(s)
- Ivo Bozovic
- Neurology Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | - Vladimir Perovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (V.P.); (D.P.); (I.V.)
| | - Ivana Basta
- Neurology Clinic, University Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (I.B.); (S.P.); (Z.S.)
| | - Stojan Peric
- Neurology Clinic, University Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (I.B.); (S.P.); (Z.S.)
| | - Zorica Stevic
- Neurology Clinic, University Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (I.B.); (S.P.); (Z.S.)
| | - Dusan Popadic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (V.P.); (D.P.); (I.V.)
| | - Irena Vukovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (V.P.); (D.P.); (I.V.)
| | | | - Emina Milosevic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (V.P.); (D.P.); (I.V.)
| |
Collapse
|
4
|
García-Fernández P, Reinhold C, Üçeyler N, Sommer C. Local Inflammatory Mediators Involved in Neuropathic Pain. Int J Mol Sci 2023; 24:ijms24097814. [PMID: 37175520 PMCID: PMC10178336 DOI: 10.3390/ijms24097814] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Polyneuropathy (PNP) is a term to describe diseases of the peripheral nervous system, 50% of which present with neuropathic pain. In some types of PNP, pain is restricted to the skin distally in the leg, suggesting a local regulatory process leading to pain. In this study, we proposed a pro-inflammatory pathway mediated by NF-κB that might be involved in the development of pain in patients with painful PNP. To test this hypothesis, we have collected nerve and skin samples from patients with different etiologies and levels of pain. We performed RT-qPCR to analyze the gene expression of the proposed inflammatory pathway components in sural nerve and in distal and proximal skin samples. In sural nerve, we showed a correlation of TLR4 and TNFα to neuropathic pain, and an upregulation of TNFα in patients with severe pain. Patients with an inflammatory PNP also presented a lower expression of TRPV1 and SIRT1. In distal skin, we found a reduced expression of TLR4 and miR-146-5p, in comparison to proximal skin. Our findings thus support our hypothesis of local inflammatory processes involved in pain in PNP, and further show disturbed anti-inflammatory pathways involving TRPV1 and SIRT1 in inflammatory PNP.
Collapse
Affiliation(s)
| | - Colette Reinhold
- Department of Neurology, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Nurcan Üçeyler
- Department of Neurology, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital of Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
5
|
Sato R, Shimizu F, Kuwahara M, Mizukami Y, Watanabe K, Maeda T, Sano Y, Takeshita Y, Koga M, Kusunoki S, Kanda T. Autocrine TNF-α Increases Penetration of Myelin-Associated Glycoprotein Antibodies Across the Blood-Nerve Barrier in Anti-MAG Neuropathy. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/3/e200086. [PMID: 36810162 PMCID: PMC9944621 DOI: 10.1212/nxi.0000000000200086] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 11/28/2022] [Indexed: 02/23/2023]
Abstract
BACKGROUND AND OBJECTIVES Deposition of myelin-associated glycoprotein (MAG) immunoglobulin M (IgM) antibodies in the sural nerve is a key feature in anti-MAG neuropathy. Whether the blood-nerve barrier (BNB) is disrupted in anti-MAG neuropathy remains elusive.We aimed to evaluate the effect of sera from anti-MAG neuropathy at the molecular level using our in vitro human BNB model and observe the change of BNB endothelial cells in the sural nerve of anti-MAG neuropathy. METHODS Diluted sera from patients with anti-MAG neuropathy (n = 16), monoclonal gammopathies of undetermined significance (MGUS) neuropathy (n = 7), amyotrophic lateral sclerosis (ALS, n = 10), and healthy controls (HCs, n = 10) incubated with human BNB endothelial cells to identify the key molecule of BNB activation using RNA-seq and a high-content imaging system, and exposed with a BNB coculture model to evaluate small molecule/IgG/IgM/anti-MAG antibody permeability. RESULTS RNA-seq and the high-content imaging system showed the significant upregulation of tumor necrosis factor (TNF-α) and nuclear factor-kappa B (NF-κB) in BNB endothelial cells after exposure to sera from patients with anti-MAG neuropathy, whereas the serum TNF-α concentration was not changed among the MAG/MGUS/ALS/HC groups. Sera from patients with anti-MAG neuropathy did not increase 10-kDa dextran or IgG permeability but enhanced IgM and anti-MAG antibody permeability. Sural nerve biopsy specimens from patients with anti-MAG neuropathy showed higher TNF-α expression levels in BNB endothelial cells and preservation of the structural integrity of the tight junctions and the presence of more vesicles in BNB endothelial cells. Neutralization of TNF-α reduces IgM/anti-MAG antibody permeability. DISCUSSION Sera from individuals with anti-MAG neuropathy increased transcellular IgM/anti-MAG antibody permeability via autocrine TNF-α secretion and NF-κB signaling in the BNB.
Collapse
Affiliation(s)
| | - Fumitaka Shimizu
- From the Department of Neurology and Clinical Neuroscience (R.S., F.S., Y.S., Y.T., Michiaki Koga, T.K.), Yamaguchi University Graduate School of Medicine, Ube; Department of Neurology (Motoi Kuwahara, S.K.), Kindai University Faculty of Medicine, Osaka; Center for Gene Research (Y.M., K.W.), Yamaguchi University, Ube, Japan; and Japan Community Health Care Organization (S.K.).
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
García-Fernández P, Höfflin K, Rausch A, Strommer K, Neumann A, Cebulla N, Reinhold AK, Rittner H, Üçeyler N, Sommer C. Systemic inflammatory markers in patients with polyneuropathies. Front Immunol 2023; 14:1067714. [PMID: 36860843 PMCID: PMC9969086 DOI: 10.3389/fimmu.2023.1067714] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction In patients with peripheral neuropathies (PNP), neuropathic pain is present in 50% of the cases, independent of the etiology. The pathophysiology of pain is poorly understood, and inflammatory processes have been found to be involved in neuro-degeneration, -regeneration and pain. While previous studies have found a local upregulation of inflammatory mediators in patients with PNP, there is a high variability described in the cytokines present systemically in sera and cerebrospinal fluid (CSF). We hypothesized that the development of PNP and neuropathic pain is associated with enhanced systemic inflammation. Methods To test our hypothesis, we performed a comprehensive analysis of the protein, lipid and gene expression of different pro- and anti-inflammatory markers in blood and CSF from patients with PNP and controls. Results While we found differences between PNP and controls in specific cytokines or lipids, such as CCL2 or oleoylcarnitine, PNP patients and controls did not present major differences in systemic inflammatory markers in general. IL-10 and CCL2 levels were related to measures of axonal damage and neuropathic pain. Lastly, we describe a strong interaction between inflammation and neurodegeneration at the nerve roots in a specific subgroup of PNP patients with blood-CSF barrier dysfunction. Conclusion In patients with PNP systemic inflammatory, markers in blood or CSF do not differ from controls in general, but specific cytokines or lipids do. Our findings further highlight the importance of CSF analysis in patients with peripheral neuropathies.
Collapse
Affiliation(s)
| | - Klemens Höfflin
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Antonia Rausch
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | | | - Astrid Neumann
- Department of Bioanalytics, Bionorica research GmbH, Innsbruck, Austria
| | - Nadine Cebulla
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Ann-Kristin Reinhold
- Department of Anesthesiology, University Hospital of Würzburg, Würzburg, Germany
| | - Heike Rittner
- Department of Anesthesiology, University Hospital of Würzburg, Würzburg, Germany
| | - Nurcan Üçeyler
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
7
|
Anti-MAG neuropathy: From biology to clinical management. J Neuroimmunol 2021; 361:577725. [PMID: 34610502 DOI: 10.1016/j.jneuroim.2021.577725] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/20/2021] [Indexed: 12/19/2022]
Abstract
The acquired chronic demyelinating neuropathies include a growing number of disease entities that have characteristic, often overlapping, clinical presentations, mediated by distinct immune mechanisms, and responding to different therapies. After the discovery in the early 1980s, that the myelin associated glycoprotein (MAG) is a target antigen in an autoimmune demyelinating neuropathy, assays to measure the presence of anti-MAG antibodies were used as the basis to diagnose the anti-MAG neuropathy. The route was open for describing the clinical characteristics of this new entity as a chronic distal large fiber sensorimotor neuropathy, for studying its pathogenesis and devising specific treatment strategies. The initial use of chemotherapeutic agents was replaced by the introduction in the late 1990s of rituximab, a monoclonal antibody against CD20+ B-cells. Since then, other anti-B cells agents have been introduced. Recently a novel antigen-specific immunotherapy neutralizing the anti-MAG antibodies with a carbohydrate-based ligand mimicking the natural HNK-1 glycoepitope has been described.
Collapse
|
8
|
Zamai L. Upregulation of the Renin-Angiotensin System Pathways and SARS-CoV-2 Infection: The Rationale for the Administration of Zinc-Chelating Agents in COVID-19 Patients. Cells 2021; 10:506. [PMID: 33673459 PMCID: PMC7997276 DOI: 10.3390/cells10030506] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
The article describes the rationale for the administration of zinc-chelating agents in COVID-19 patients. In a previous work I have highlighted that the binding of the SARS-CoV spike proteins to the zinc-metalloprotease ACE2 has been shown to induce ACE2 shedding by activating the zinc-metalloprotease ADAM17, which ultimately leads to systemic upregulation of ACE2 activity. Moreover, based on experimental models, it was also shown the detrimental effect of the excessive systemic activity of ACE2 through its downstream pathways, which leads to "clinical" manifestations resembling COVID-19. In this regard, strong upregulation of circulating ACE2 activity was recently reported in COVID-19 patients, thus supporting the previous hypothesis that COVID-19 may derive from upregulation of ACE2 activity. Based on this, a reasonable hypothesis of using inhibitors that curb the upregulation of both ACE2 and ADAM17 zinc-metalloprotease activities and consequent positive feedback-loops (initially triggered by SARS-CoV-2 and subsequently sustained independently on viral trigger) is proposed as therapy for COVID-19. In particular, zinc-chelating agents such as citrate and ethylenediaminetetraacetic acid (EDTA) alone or in combination are expected to act in protecting from COVID-19 at different levels thanks to their both anticoagulant properties and inhibitory activity on zinc-metalloproteases. Several arguments are presented in support of this hypothesis and based on the current knowledge of both beneficial/harmful effects and cost/effectiveness, the use of chelating agents in the prevention and therapy of COVID-19 is proposed. In this regard, clinical trials (currently absent) employing citrate/EDTA in COVID-19 are urgently needed in order to shed more light on the efficacy of zinc chelators against SARS-CoV-2 infection in vivo.
Collapse
Affiliation(s)
- Loris Zamai
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy;
- National Institute for Nuclear Physics (INFN)-Gran Sasso National Laboratory (LNGS), Assergi, 67100 L’Aquila, Italy
| |
Collapse
|