1
|
Zhou J, Yang X, Wang X, Li B, Xu Y, Zhang H, Zhu Y, Wang X, Feng J. Immunotherapy for neuromyelitis optica spectrum disorder: a comparative analysis of efficacy and safety of azathioprine, mycophenolate mofetil, tacrolimus, and rituximab. Front Neurol 2025; 16:1559118. [PMID: 40356634 PMCID: PMC12066257 DOI: 10.3389/fneur.2025.1559118] [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: 01/11/2025] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Background and purpose Biologic therapies are anticipated to dominate the treatment landscape for neuromyelitis optica spectrum disorders (NMOSD) in the future. Despite this, many patients in China continue to use off-label medications due to economic and other constraints. A multicenter NMOSD cohort study was conducted to compare the efficacy and safety of tacrolimus (TAC), mycophenolate mofetil (MMF), azathioprine (AZA), and rituximab (RTX). The objective of this study is to provide a clinical evidence-based reference for patients who still require the use of these off-label medications. Methods This retrospective study included NMOSD patients treated with TAC (n = 24), MMF (n = 74), AZA (n = 34), and RTX (n = 81). Of these, 81 underwent magnetic resonance imaging (MRI) activity analysis during follow-up. The observation period commenced with the treatment initiation and extended until August 31, 2023. The primary efficacy outcome was the time to the first relapse post-immunotherapy initiation, The hazard ratio (HR) was analyzed using the Cox proportional hazards model to compare the relative risk of the first relapse between different treatment groups (e.g., RTX, MMF, TAC, and AZA). Secondary outcomes encompassed annualized relapse rate (ARR), MRI activity, drug persistence, and relapse rate (RR). The safety outcome was the occurrence of severe adverse drug reaction events. Results A total of 213 patients were included in the study. During the first year of immunotherapy, patients treated with RTX (HR = 18.41, 95% CI: 4.039-83.87; p < 0.05) and MMF (HR = 22.72, 95% CI: 4.783-108.0; p < 0.0001) experienced a significantly lower risk of relapse compared to those treated with tacrolimus (TAC). The risk of first relapse in the AZA group was higher compared to the RTX group (HR = 2.786, 95% CI: 0.4771-16.27; p = 0.2551) and the MMF group (HR = 4.005, 95% CI: 0.5973-26.86; p = 0.1529), although the differences were not statistically significant. In the second year, this trend continued with RTX (HR = 6.200, 95% CI: 1.825-21.06; p = 0.0034) and MMF (HR = 6.017, 95% CI: 1.782-20.32; p = 0.0039) demonstrating a lower relapse risk compared to oral TAC. Similarly, RTX and MMF were more effective than oral AZA in reducing relapse risk (RTX: HR = 3.510, 95% CI: 1.202-10.25; p = 0.0216; MMF: HR = 3.909, 95% CI: 1.318-11.59; p = 0.0140). The difference in the risk of the first relapse between the MMF and RTX groups was not statistically significant (HR = 0.7217, p = 0.7156 in the first year; HR = 0.9351, p = 0.9003 in the second year) although the difference was not statistically significant. The risk of first relapse was higher in the group treated with oral conventional immunosuppressants (ISTs) compared to the RTX group, (HR = 2.170, p = 0.1449 in the first year; HR = 1.820, p = 0.1091 in the second year). The annual relapse rate (ARR) significantly decreased after treatment with all four drugs. RTX and MMF were more effective in controlling disease relapse compared to TAC and AZA, though these differences were not statistically significant (RTX: ARR = 0.12, 95% CI: 0.03-0.21; MMF: ARR = 0.15, 95% CI: 0.07-0.23; TAC: ARR = 0.21, 95% CI: 0.03-0.39; AZA: ARR = 0.19, 95% CI: 0.08-0.3; p = 0.81). When combining clinical and relapse-independent MRI activity analyses in 81 NMOSD patients, RTX demonstrated superior control of disease activity, with a statistically significant difference (p = 0.036). No hospitalization events related to severe drug adverse effects were reported in either the IST or RTX groups. Conclusion The study provides data comparing the efficacy of various off-label treatments in a Chinese NMOSD cohort, illustrating that RTX is more effective than traditional immunosuppressants in controlling NMOSD relapses and disease activity but no superiority in the time to the first relapse post-immunotherapy initiation. RTX and MMF may offer superior treatment alternatives for NMOSD patients compared to TAC and AZA.
Collapse
Affiliation(s)
- Jing Zhou
- Jinan University, Guangzhou, Guangdong, China
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiaolin Yang
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyi Wang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
| | - Bin Li
- First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun Xu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | | | - Yinxin Zhu
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiaoming Wang
- Jinan University, Guangzhou, Guangdong, China
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jinzhou Feng
- Department of Neurology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Sánchez-Sanz A, Muñoz-Viana R, Sabín-Muñoz J, Moreno-Torres I, Brea-Álvarez B, Rodríguez-De la Fuente O, García-Merino A, Sánchez-López AJ. Response to Fingolimod in Multiple Sclerosis Patients Is Associated with a Differential Transcriptomic Regulation. Int J Mol Sci 2024; 25:1372. [PMID: 38338652 PMCID: PMC10855583 DOI: 10.3390/ijms25031372] [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: 12/22/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Fingolimod is an immunomodulatory sphingosine-1-phosphate (S1P) analogue approved for the treatment of relapsing-remitting multiple sclerosis (RRMS). The identification of biomarkers of clinical responses to fingolimod is a major necessity in MS to identify optimal responders and avoid the risk of disease progression in non-responders. With this aim, we used RNA sequencing to study the transcriptomic changes induced by fingolimod in peripheral blood mononuclear cells of MS-treated patients and their association with clinical response. Samples were obtained from 10 RRMS patients (five responders and five non-responders) at baseline and at 12 months of fingolimod therapy. Fingolimod exerted a vast impact at the transcriptional level, identifying 7155 differentially expressed genes (DEGs) compared to baseline that affected the regulation of numerous signaling pathways. These DEGs were predominantly immune related, including genes associated with S1P metabolism, cytokines, lymphocyte trafficking, master transcription factors of lymphocyte functions and the NF-kB pathway. Responder and non-responder patients exhibited a differential transcriptomic regulation during treatment, with responders presenting a higher number of DEGs (6405) compared to non-responders (2653). The S1P, NF-kB and TCR signaling pathways were differentially modulated in responder and non-responder patients. These transcriptomic differences offer the potential of being exploited as biomarkers of a clinical response to fingolimod.
Collapse
Affiliation(s)
- Alicia Sánchez-Sanz
- Neuroimmunology Unit, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain;
| | - Rafael Muñoz-Viana
- Bioinformatics Unit, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain;
| | - Julia Sabín-Muñoz
- Department of Neurology, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Madrid, Spain; (J.S.-M.); (O.R.-D.l.F.)
| | - Irene Moreno-Torres
- Demyelinating Diseases Unit, Hospital Universitario Fundación Jiménez Díaz, 28040 Madrid, Spain;
| | - Beatriz Brea-Álvarez
- Radiodiagnostic Division, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Madrid, Spain;
| | - Ofir Rodríguez-De la Fuente
- Department of Neurology, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Madrid, Spain; (J.S.-M.); (O.R.-D.l.F.)
| | - Antonio García-Merino
- Neuroimmunology Unit, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain;
- Department of Neurology, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Madrid, Spain; (J.S.-M.); (O.R.-D.l.F.)
- Department of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Red Española de Esclerosis Múltiple (REEM), 08028 Barcelona, Spain
| | - Antonio J. Sánchez-López
- Neuroimmunology Unit, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain;
- Red Española de Esclerosis Múltiple (REEM), 08028 Barcelona, Spain
- Biobank, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain
| |
Collapse
|
3
|
Ruggieri S, Prosperini L, Al-Araji S, Annovazzi PO, Bisecco A, Ciccarelli O, De Stefano N, Filippi M, Fleischer V, Evangelou N, Enzinger C, Gallo A, Garjani A, Groppa S, Haggiag S, Khalil M, Lucchini M, Mirabella M, Montalban X, Pozzilli C, Preziosa P, Río J, Rocca MA, Rovira A, Stromillo ML, Zaffaroni M, Tortorella C, Gasperini C. Assessing treatment response to oral drugs for multiple sclerosis in real-world setting: a MAGNIMS Study. J Neurol Neurosurg Psychiatry 2024; 95:142-150. [PMID: 37775266 DOI: 10.1136/jnnp-2023-331920] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/09/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND The assessment of treatment response is a crucial step for patients with relapsing-remitting multiple sclerosis on disease-modifying therapies (DMTs). We explored whether a scoring system developed within the MAGNIMS (MRI in Multiple Sclerosis) network to evaluate treatment response to injectable drugs can be adopted also to oral DMTs. METHODS A multicentre dataset of 1200 patients who started three oral DMTs (fingolimod, teriflunomide and dimethyl fumarate) was collected within the MAGNIMS network. Disease activity after the first year was classified by the 'MAGNIMS' score based on the combination of relapses (0-≥2) and/or new T2 lesions (<3 or ≥3) on brain MRI. We explored the association of this score with the following 3-year outcomes: (1) confirmed disability worsening (CDW); (2) treatment failure (TFL); (3) relapse count between years 1 and 3. The additional value of contrast-enhancing lesions (CELs) and lesion location was explored. RESULTS At 3 years, 160 patients experienced CDW: 12% of them scored '0' (reference), 18% scored '1' (HR=1.82, 95% CI 1.20 to 2.76, p=0.005) and 37% scored '2' (HR=2.74, 95% CI 1.41 to 5.36, p=0.003) at 1 year. The analysis of other outcomes provided similar findings. Considering the location of new T2 lesions (supratentorial vs infratentorial/spinal cord) and the presence of CELs improved the prediction of CDW and TFL, respectively, in patients with minimal MRI activity alone (one or two new T2 lesions). CONCLUSIONS Early relapses and substantial MRI activity in the first year of treatment are associated with worse short-term outcomes in patients treated with some of the oral DMTs.
Collapse
Affiliation(s)
- Serena Ruggieri
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Luca Prosperini
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | - Sarmad Al-Araji
- Department of Neuroinflammation, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Pietro Osvaldo Annovazzi
- Neuroimmunology Unit-Multiple Sclerosis Center, Hospital of Gallarate, ASST della Valle Olona, Gallarate, Italy
| | - Alvino Bisecco
- Department of Advanced Medical and Surgical Sciences, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Olga Ciccarelli
- Department of Neuroinflammation, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
- National Institute for Health Research Biomedical Research Centre, University College London Hospitals, London, UK
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Massimo Filippi
- Neurology Unit and Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Vinzenz Fleischer
- Department of Neurology and Neuroimaging Center (NIC) of the Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Nikos Evangelou
- Mental Health & Clinical Neuroscience Unit, University of Nottingham, Nottingham, UK
- Department of Neurology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Christian Enzinger
- Department of Neurology, Medical University of Graz, Graz, Austria
- Department of Radiology (Division of Neuroradiology, Vascular and Interventional Radiology), Medical University of Graz, Graz, Austria
| | - Antonio Gallo
- Department of Advanced Medical and Surgical Sciences, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Afagh Garjani
- Mental Health & Clinical Neuroscience Unit, University of Nottingham, Nottingham, UK
- Department of Neurology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Sergiu Groppa
- Department of Neurology and Neuroimaging Center (NIC) of the Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Shalom Haggiag
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | - Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Matteo Lucchini
- Multiple Sclerosis Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Centro di ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Rome, Italy
| | - Massimiliano Mirabella
- Multiple Sclerosis Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Centro di ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Rome, Italy
| | - Xavier Montalban
- Centre d'Esclerosi Multiple de Catalunya (Cemcat), Department of Neurology/Neuroimmunology, Hospital Universitari Vall d'Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Carlo Pozzilli
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Paolo Preziosa
- Neurology Unit and Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Jordi Río
- Centre d'Esclerosi Multiple de Catalunya (Cemcat), Department of Neurology/Neuroimmunology, Hospital Universitari Vall d'Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Maria A Rocca
- Neurology Unit and Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alex Rovira
- Section of Neuroradiology, Department of Radiology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria L Stromillo
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Mauro Zaffaroni
- Neuroimmunology Unit-Multiple Sclerosis Center, Hospital of Gallarate, ASST della Valle Olona, Gallarate, Italy
| | - Carla Tortorella
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| | - Claudio Gasperini
- Department of Neurosciences, San Camillo Forlanini Hospital, Rome, Italy
| |
Collapse
|
4
|
Vakrakou AG, Brinia ME, Alexaki A, Koumasopoulos E, Stathopoulos P, Evangelopoulos ME, Stefanis L, Stadelmann-Nessler C, Kilidireas C. Multiple faces of multiple sclerosis in the era of highly efficient treatment modalities: Lymphopenia and switching treatment options challenges daily practice. Int Immunopharmacol 2023; 125:111192. [PMID: 37951198 DOI: 10.1016/j.intimp.2023.111192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/13/2023]
Abstract
The expanded treatment landscape in relapsing-remitting multiple sclerosis (MS) has resulted in highly effective treatment options and complexity in managing disease- or drug-related events during disease progression. Proper decision-making requires thorough knowledge of the immunobiology of MS itself and an understanding of the main principles behind the mechanisms that lead to secondary autoimmunity affecting organs other than the central nervous system as well as opportunistic infections. The immune system is highly adapted to both environmental and disease-modifying agents. Immune reconstitution following cell depletion or cell entrapment therapies eliminates pathogenic aspects of the disease but can also lead to distorted immune responses with harmful effects. Atypical relapses occur with second-line treatments or after their discontinuation and require appropriate clinical decisions. Lymphopenia is a result of the mechanism of action of many drugs used to treat MS. However, persistent lymphopenia and cell-specific lymphopenia could result in disease exacerbation, secondary autoimmunity, or the emergence of opportunistic infections. Clinicians treating patients with MS should be aware of the multiple faces of MS under novel, efficient treatment modalities and understand the intricate brain-immune cell interactions in the context of an altered immune system. MS relapses and disease progression still occur despite the current treatment modalities and are mediated either by failure to control effector mechanisms inherent to MS pathophysiology or by new drug-related mechanisms. The multiple faces of MS due to the highly adapted immune system of patients impose the need for appropriate switching therapies that safeguard disease remission and further clinical improvement.
Collapse
Affiliation(s)
- Aigli G Vakrakou
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Department of Neuropathology, University of Göttingen Medical Center, Göttingen, Germany.
| | - Maria-Evgenia Brinia
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Alexaki
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Koumasopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panos Stathopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Eleftheria Evangelopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Leonidas Stefanis
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Constantinos Kilidireas
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aiginition Hospital, National and Kapodistrian University of Athens, Athens, Greece; Department of Neurology, Henry Dunant Hospital Center, Athens, Greece
| |
Collapse
|
5
|
Gonzalez-Martinez A, Patel R, Healy BC, Lokhande H, Paul A, Saxena S, Polgar-Turcsanyi M, Weiner HL, Chitnis T. miRNA 548a-3p as biomarker of NEDA-3 at 2 years in multiple sclerosis patients treated with fingolimod. J Neuroinflammation 2023; 20:131. [PMID: 37254147 DOI: 10.1186/s12974-023-02811-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/19/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a disabling autoimmune demyelinating disorder affecting young people and causing significant disability. In the last decade, different microRNA (miRNA) expression patterns have been associated to several treatment response therapies such as interferon and glatiramer acetate. Nowadays, there is increasing interest in the potential role of miRNA as treatment response biomarkers to the most recent oral and intravenous treatments. In this study, we aimed to evaluate serum miRNAs as biomarkers of No Evidence of Disease Activity (NEDA-3) at 2 years in patients with relapsing remitting MS (RRMS) treated with fingolimod. MAIN BODY A Discovery cohort of 31 RRMS patients treated with fingolimod were identified from the CLIMB study and classified as No Evidence of Disease Activity (NEDA-3) or Evidence of Disease Activity (EDA-3) after 2 years on treatment. Levels of miRNA expression were measured at 6 months using human serum miRNA panels and compared in EDA-3 and NEDA-3 groups using the Wilcoxon rank sum test. A set of differentially expressed miRNA was further validated in an independent cohort of 22 fingolimod-treated patients. We found that 548a-3p serum levels were higher levels in fingolimod-treated patients classified as NEDA-3, compared to the EDA-3 group in both the Discovery (n = 31; p = 0.04) and Validation (n = 22; p = 0.03) cohorts 6 months after treatment initiation; miR-548a-3p provided an AUC of 0.882 discriminating patients with NEDA-3 at 2 years in the Validation cohort. CONCLUSION Our results show differences in miR-548a-3p expression at 6 months after fingolimod start in patients with MS with NEDA-3 at 2 years. These results provide class III evidence of the use of miR-548a-3p as biomarker of NEDA-3 in patients with fingolimod.
Collapse
Affiliation(s)
- Alicia Gonzalez-Martinez
- Department of Neurology, Translational Neuroimmunology Research Center (TNRC), Ann Romney Center for Neurologic Diseases (ARCND), Brigham and Women's Hospital, 60 Fenwood Road, 9002K, Boston, MA, 02115, USA
| | - Rohit Patel
- Department of Neurology, Translational Neuroimmunology Research Center (TNRC), Ann Romney Center for Neurologic Diseases (ARCND), Brigham and Women's Hospital, 60 Fenwood Road, 9002K, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Brian C Healy
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurology, Brigham MS Center, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Hrishikesh Lokhande
- Department of Neurology, Translational Neuroimmunology Research Center (TNRC), Ann Romney Center for Neurologic Diseases (ARCND), Brigham and Women's Hospital, 60 Fenwood Road, 9002K, Boston, MA, 02115, USA
| | - Anu Paul
- Department of Neurology, Translational Neuroimmunology Research Center (TNRC), Ann Romney Center for Neurologic Diseases (ARCND), Brigham and Women's Hospital, 60 Fenwood Road, 9002K, Boston, MA, 02115, USA
| | - Shrishti Saxena
- Department of Neurology, Translational Neuroimmunology Research Center (TNRC), Ann Romney Center for Neurologic Diseases (ARCND), Brigham and Women's Hospital, 60 Fenwood Road, 9002K, Boston, MA, 02115, USA
| | - Mariann Polgar-Turcsanyi
- Department of Neurology, Translational Neuroimmunology Research Center (TNRC), Ann Romney Center for Neurologic Diseases (ARCND), Brigham and Women's Hospital, 60 Fenwood Road, 9002K, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Howard L Weiner
- Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurology, Brigham MS Center, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Tanuja Chitnis
- Department of Neurology, Translational Neuroimmunology Research Center (TNRC), Ann Romney Center for Neurologic Diseases (ARCND), Brigham and Women's Hospital, 60 Fenwood Road, 9002K, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
- Department of Neurology, Brigham MS Center, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
6
|
Malhotra S, Hurtado-Navarro L, Pappolla A, Villar LMM, Río J, Montalban X, Pelegrin P, Comabella M. Increased NLRP3 Inflammasome Activation and Pyroptosis in Patients With Multiple Sclerosis With Fingolimod Treatment Failure. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/3/e200100. [PMID: 36973075 PMCID: PMC10042441 DOI: 10.1212/nxi.0000000000200100] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/12/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND AND OBJECTIVES Inflammasomes are involved in the pathogenesis of different neuroimmune and neurodegenerative diseases, including multiple sclerosis (MS). In a previous study by our group, the nucleotide-binding oligomerization domain, leucine-rich repeat receptor and pyrin-domain-containing 3 (NLRP3) inflammasome was reported to be associated with the response to interferon-beta in MS. Based on recent data showing the potential for the oral therapy fingolimod to inhibit NLRP3 inflammasome activation, here we investigated whether fingolimod could also be implicated in the response to this therapy in patients with MS. METHODS NLRP3 gene expression levels were measured by real-time PCR in peripheral blood mononuclear cells at baseline and after 3, 6, and 12 months in a cohort of patients with MS treated with fingolimod (N = 23), dimethyl fumarate (N = 21), and teriflunomide (N = 21) and classified into responders and nonresponders to the treatment according to clinical and radiologic criteria. In a subgroup of fingolimod responders and nonresponders, the percentage of monocytes with an oligomer of ASC was determined by flow cytometry, and the levels of interleukin (IL)-1β, IL-18, IL-6, tumor necrosis factor (TNF)α, and galectin-3 were quantified by ELISA. RESULTS NLPR3 expression levels were significantly increased in fingolimod nonresponders after 3 (p = 0.03) and 6 months (p = 0.008) of treatment compared with the baseline but remained similar in responders at all time points. These changes were not observed in nonresponders to the other oral therapies tested. The formation of an oligomer of ASC in monocytes after lipopolysaccharide and adenosine 5'-triphosphate stimulation was significantly decreased in responders (p = 0.006) but increased in nonresponders (p = 0.0003) after 6 months of fingolimod treatment compared with the baseline. Proinflammatory cytokine release from stimulated peripheral blood mononuclear cells was comparable between responders and nonresponders, but galectin-3 levels on cell supernatants, as a marker of cell damage, were significantly increased in fingolimod nonresponders (p = 0.02). DISCUSSION The differential effect of fingolimod on the formation of an inflammasome-triggered ASC oligomer in monocytes between responders and nonresponders could be used as a response biomarker after 6 months of fingolimod treatment and suggests that fingolimod may exert their beneficial effects by reducing inflammasome signaling in a subset of patients with MS.
Collapse
Affiliation(s)
- Sunny Malhotra
- From the Servei de Neurologia-Neuroimmunologia (S.M., A.P., J.R., X.M., M.C.), Centre d´Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d´Hebron (VHIR), Hospital Universitari Vall d´Hebron, Universitat Autònoma de Barcelona, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca) (L.H.-N., P.P.), University Clinical Hospital Virgen de la Arrixaca, Spain; Departments of Neurology and Immunology (L.M.M.V.), Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain; and Department of Biochemistry and Molecular Biology B and Immunology (P.P.), Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Laura Hurtado-Navarro
- From the Servei de Neurologia-Neuroimmunologia (S.M., A.P., J.R., X.M., M.C.), Centre d´Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d´Hebron (VHIR), Hospital Universitari Vall d´Hebron, Universitat Autònoma de Barcelona, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca) (L.H.-N., P.P.), University Clinical Hospital Virgen de la Arrixaca, Spain; Departments of Neurology and Immunology (L.M.M.V.), Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain; and Department of Biochemistry and Molecular Biology B and Immunology (P.P.), Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Agustin Pappolla
- From the Servei de Neurologia-Neuroimmunologia (S.M., A.P., J.R., X.M., M.C.), Centre d´Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d´Hebron (VHIR), Hospital Universitari Vall d´Hebron, Universitat Autònoma de Barcelona, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca) (L.H.-N., P.P.), University Clinical Hospital Virgen de la Arrixaca, Spain; Departments of Neurology and Immunology (L.M.M.V.), Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain; and Department of Biochemistry and Molecular Biology B and Immunology (P.P.), Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Luisa M M Villar
- From the Servei de Neurologia-Neuroimmunologia (S.M., A.P., J.R., X.M., M.C.), Centre d´Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d´Hebron (VHIR), Hospital Universitari Vall d´Hebron, Universitat Autònoma de Barcelona, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca) (L.H.-N., P.P.), University Clinical Hospital Virgen de la Arrixaca, Spain; Departments of Neurology and Immunology (L.M.M.V.), Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain; and Department of Biochemistry and Molecular Biology B and Immunology (P.P.), Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Jordi Río
- From the Servei de Neurologia-Neuroimmunologia (S.M., A.P., J.R., X.M., M.C.), Centre d´Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d´Hebron (VHIR), Hospital Universitari Vall d´Hebron, Universitat Autònoma de Barcelona, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca) (L.H.-N., P.P.), University Clinical Hospital Virgen de la Arrixaca, Spain; Departments of Neurology and Immunology (L.M.M.V.), Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain; and Department of Biochemistry and Molecular Biology B and Immunology (P.P.), Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Xavier Montalban
- From the Servei de Neurologia-Neuroimmunologia (S.M., A.P., J.R., X.M., M.C.), Centre d´Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d´Hebron (VHIR), Hospital Universitari Vall d´Hebron, Universitat Autònoma de Barcelona, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca) (L.H.-N., P.P.), University Clinical Hospital Virgen de la Arrixaca, Spain; Departments of Neurology and Immunology (L.M.M.V.), Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain; and Department of Biochemistry and Molecular Biology B and Immunology (P.P.), Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Pablo Pelegrin
- From the Servei de Neurologia-Neuroimmunologia (S.M., A.P., J.R., X.M., M.C.), Centre d´Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d´Hebron (VHIR), Hospital Universitari Vall d´Hebron, Universitat Autònoma de Barcelona, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca) (L.H.-N., P.P.), University Clinical Hospital Virgen de la Arrixaca, Spain; Departments of Neurology and Immunology (L.M.M.V.), Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain; and Department of Biochemistry and Molecular Biology B and Immunology (P.P.), Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Manuel Comabella
- From the Servei de Neurologia-Neuroimmunologia (S.M., A.P., J.R., X.M., M.C.), Centre d´Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d´Hebron (VHIR), Hospital Universitari Vall d´Hebron, Universitat Autònoma de Barcelona, Spain; Biomedical Research Institute of Murcia (IMIB-Arrixaca) (L.H.-N., P.P.), University Clinical Hospital Virgen de la Arrixaca, Spain; Departments of Neurology and Immunology (L.M.M.V.), Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigacion Sanitaria, Madrid, Spain; and Department of Biochemistry and Molecular Biology B and Immunology (P.P.), Faculty of Medicine, University of Murcia, Murcia, Spain.
| |
Collapse
|
7
|
Xu QQ, Zhang XL, Luo B, Li T, Yuan G, Shao SY. Role of Peripheral Regulatory T Lymphocytes in Patients with Thyroid Associated Ophthalmopathy During Systemic Glucocorticoid Treatment: A Prospective Observational Study. Curr Med Sci 2023; 43:130-138. [PMID: 36459302 DOI: 10.1007/s11596-022-2671-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/31/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVE Thyroid-associated ophthalmopathy (TAO) is an autoimmune disorder involving the orbital tissue. This study aimed to understand the role of regulatory T cells (Tregs) in TAO during 12-week systemic glucocorticoid (GC) treatment. METHODS Thirty-two moderate-severe TAO patients with a clinical activity score (CAS) ≥3/7 or with prolonged T2 relaxation time (T2RT) on at least one side of extraocular muscle (EOM) were enrolled. The percentage of the peripheral CD4+CD25(high)CD127(-/low) Tregs was analyzed using flow cytometry before and after the GC treatment. The activity and severity of TAO, T2RT, and the clinical outcomes after the GC treatment were assessed. Their correlation with the peripheral Tregs was investigated. RESULTS There was no significant association between the baseline Treg fraction and the activity and severity of TAO or the treatment response. A significant reduction of Tregs was observed after the GC therapy merely in patients without any clinical improvement. CONCLUSION Treg reduction after systemic GC therapy is indicative of a poor therapeutic response. Accordingly, dynamic alterations of Tregs could help to evaluate the effectiveness of the GC treatment.
Collapse
Affiliation(s)
- Qin-Qin Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Branch of the National Clinical Research Center for Metabolic Diseases, Wuhan, 430030, China
| | - Xiao-Ling Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Branch of the National Clinical Research Center for Metabolic Diseases, Wuhan, 430030, China
| | - Ban Luo
- Depatment of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tao Li
- Depatment of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gang Yuan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Branch of the National Clinical Research Center for Metabolic Diseases, Wuhan, 430030, China
| | - Shi-Ying Shao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Branch of the National Clinical Research Center for Metabolic Diseases, Wuhan, 430030, China.
| |
Collapse
|
8
|
Camacho-Toledano C, Machín-Díaz I, Calahorra L, Cabañas-Cotillas M, Otaegui D, Castillo-Triviño T, Villar LM, Costa-Frossard L, Comabella M, Midaglia L, García-Domínguez JM, García-Arocha J, Ortega MC, Clemente D. Peripheral myeloid-derived suppressor cells are good biomarkers of the efficacy of fingolimod in multiple sclerosis. J Neuroinflammation 2022; 19:277. [PMCID: PMC9675277 DOI: 10.1186/s12974-022-02635-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022] Open
Abstract
Background The increasing number of treatments that are now available to manage patients with multiple sclerosis (MS) highlights the need to develop biomarkers that can be used within the framework of individualized medicine. Fingolimod is a disease-modifying treatment that belongs to the sphingosine-1-phosphate receptor modulators. In addition to inhibiting T cell egress from lymph nodes, fingolimod promotes the immunosuppressive activity of myeloid-derived suppressor cells (MDSCs), whose monocytic subset (M-MDSCs) can be used as a biomarker of disease severity, as well as the degree of demyelination and extent of axonal damage in the experimental autoimmune encephalomyelitis (EAE) model of MS. In the present study, we have assessed whether the abundance of circulating M-MDSCs may represent a useful biomarker of fingolimod efficacy in EAE and in the clinical context of MS patients. Methods Treatment with vehicle or fingolimod was orally administered to EAE mice for 14 days in an individualized manner, starting the day when each mouse began to develop clinical signs. Peripheral blood from EAE mice was collected previous to treatment and human peripheral blood mononuclear cells (PBMCs) were collected from fingolimod to treat MS patients’ peripheral blood. In both cases, M-MDSCs abundance was analyzed by flow cytometry and its relationship with the future clinical affectation of each individual animal or patient was assessed. Results Fingolimod-treated animals presented a milder EAE course with less demyelination and axonal damage, although a few animals did not respond well to treatment and they invariably had fewer M-MDSCs prior to initiating the treatment. Remarkably, M-MDSC abundance was also found to be an important and specific parameter to distinguish EAE mice prone to better fingolimod efficacy. Finally, in a translational effort, M-MDSCs were quantified in MS patients at baseline and correlated with different clinical parameters after 12 months of fingolimod treatment. M-MDSCs at baseline were highly representative of a good therapeutic response to fingolimod, i.e., patients who met at least two of the criteria used to define non-evidence of disease activity-3 (NEDA-3) 12 months after treatment. Conclusion Our data indicate that M-MDSCs might be a useful predictive biomarker of the response of MS patients to fingolimod. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02635-3.
Collapse
Affiliation(s)
- Celia Camacho-Toledano
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Isabel Machín-Díaz
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Leticia Calahorra
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - María Cabañas-Cotillas
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - David Otaegui
- grid.432380.eMultiple Sclerosis Unit, Biodonostia Health Institute, 20014 Donostia-San Sebastián, Spain
| | - Tamara Castillo-Triviño
- grid.432380.eMultiple Sclerosis Unit, Biodonostia Health Institute, 20014 Donostia-San Sebastián, Spain ,grid.414651.30000 0000 9920 5292Neurology Department, Hospital Universitario Donostia, San Sebastián, Spain
| | - Luisa María Villar
- grid.411347.40000 0000 9248 5770Immunology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Lucienne Costa-Frossard
- grid.411347.40000 0000 9248 5770Immunology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain ,grid.411347.40000 0000 9248 5770Multiple Sclerosis Unit, Neurology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Manuel Comabella
- grid.411083.f0000 0001 0675 8654Neurology-Neuroimmunology Service, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Luciana Midaglia
- grid.411083.f0000 0001 0675 8654Neurology-Neuroimmunology Service, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José Manuel García-Domínguez
- grid.410526.40000 0001 0277 7938Multiple Sclerosis Unit, Department of Neurology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Jennifer García-Arocha
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - María Cristina Ortega
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Diego Clemente
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW Increasingly, therapeutic strategy in multiple sclerosis (MS) is informed by imaging and laboratory biomarkers, in addition to traditional clinical factors. Here, we review aspects of monitoring the efficacy and risks of disease-modifying therapy (DMT) with both conventional and emerging MRI and laboratory measures. RECENT FINDINGS The adoption of consensus-driven, stable MRI acquisition protocols and artificial intelligence-based, quantitative image analysis is heralding an era of precision monitoring of DMT efficacy. New MRI measures of compartmentalized inflammation, neuro-degeneration and repair complement traditional metrics but require validation before use in individual patients. Laboratory markers of brain cellular injury, such as neurofilament light, are robust outcomes in DMT efficacy trials; their use in clinical practice is being refined. DMT-specific laboratory monitoring for safety is critical and may include lymphocytes, immunoglobulins, autoimmunity surveillance, John Cunningham virus serology and COVID-19 vaccination seroresponse. SUMMARY A biomarker-enhanced monitoring strategy has immediate clinical application, with growing evidence of long-term reductions in disability accrual when both clinically symptomatic and asymptomatic inflammatory activity is fully suppressed; and amelioration of the risks associated with therapy. Emerging MRI and blood-based measures will also become important tools for monitoring agents that target the innate immune system and promote neuro-repair.
Collapse
|
10
|
Papeix C, Castelnovo G, Leray E, Coustans M, Levy P, Visy JM, Kobelt G, Lamy F, Allaf B, Heintzmann F, Chouette I, Raponi E, Durand B, Grevat E, Kamar D, Debouverie M, Lebrun-Frenay C. Long-Term Effectiveness, Safety and Tolerability of Fingolimod in Patients with Multiple Sclerosis in Real-World Treatment Settings in France: The VIRGILE Study. Neurol Ther 2022; 11:633-658. [PMID: 35147904 PMCID: PMC9095796 DOI: 10.1007/s40120-022-00334-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 01/28/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction It is important to confirm the effectiveness and tolerability of disease-modifying treatments for relapsing–remitting multiple sclerosis (RRMS) in real-world treatment settings. This prospective observational cohort study (VIRGILE) was performed at the request of the French health authorities. The primary objective was to evaluate the effectiveness of fingolimod 0.5 mg in reducing the annualised relapse rate (ARR) in patients with RRMS. Methods Participating neurologists enrolled all adult patients with RRMS starting fingolimod treatment between 2014 and 2016, who were followed for 3 years. Follow-up consultations took place at the investigator’s discretion. The primary outcome measure was the change in ARR at month 24 after fingolimod initiation. Relapses and adverse events were documented at each consultation; disability assessment (EDSS) and magnetic resonance imagery were performed at the investigator’s discretion. Results Of 1055 eligible patients, 633 patients were assessable at month 36; 405 (64.0%) were treated continuously with fingolimod for 3 years. The ARR decreased from 0.92 ± 0.92 at inclusion to 0.31 ± 0.51 at month 24, a significant reduction of 0.58 [95% CI − 0.51 to − 0.65] relapses/year (p < 0.001). Since starting fingolimod, 461 patients (60.9%) remained relapse-free at month 24 and 366 patients (55.5%) at month 36. In multivariate analysis, no previous disease-modifying treatment, number of relapses in the previous year and lower EDSS score at inclusion were associated with a greater on-treatment reduction in ARR. The mean EDSS score remained stable over the course of the study. Sixty-one out of 289 (21.1%) patients presented new radiological signs of disease activity. Treatment-related serious adverse events were lymphopenia (N = 21), bradycardia (N = 19), elevated transaminases (N = 9) and macular oedema (N = 9). Conclusions The effectiveness and tolerability of fingolimod in everyday clinical practice are consistent with findings of previous phase III studies. Our study highlights the utility of fingolimod for the long-term management of patients with multiple sclerosis. Supplementary Information The online version contains supplementary material available at 10.1007/s40120-022-00334-y.
Collapse
Affiliation(s)
- Caroline Papeix
- Département de Neurologie, Hôpital de la Pitié Salpêtrière, APHP 6, 83 Boulevard de l'Hôpital, 75013, Paris, France.
| | | | | | - Marc Coustans
- Service de Neurologie, Hôpital Laënnec, Quimper, France
| | - Pierre Levy
- LEDa, LEGOS, Université Paris-Dauphine, PSL Research University, Paris, France
| | | | | | | | | | | | | | - Eric Raponi
- Novartis Pharma S.A.S., Rueil-Malmaison, France
| | | | | | - Driss Kamar
- Ividata Life Science, Levallois-Perret, France
| | | | | | | |
Collapse
|
11
|
Huh SY, Kim SH, Kim KH, Kwon YN, Kim SM, Kim SW, Shin HY, Chung YH, Min JH, So J, Lim YM, Kim KK, Kim NH, Nam TS, Kang SY, Oh J, Oh SI, Sohn E, Kim HJ. Safety and Temporal Pattern of the Lymphocyte Count During Fingolimod Therapy in Patients With Multiple Sclerosis: Real-World Korean Experience. J Clin Neurol 2022; 18:663-670. [DOI: 10.3988/jcn.2022.18.6.663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/17/2022] [Accepted: 03/17/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- So-Young Huh
- Department of Neurology, College of Medicine, Kosin University, Busan, Korea
| | - Su-Hyun Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Ki Hoon Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Young Nam Kwon
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - Sung-Min Kim
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - Seung Woo Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Ha Young Shin
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Yeon Hak Chung
- Department of Neurology, Neuroscience Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ju-Hong Min
- Department of Neurology, Neuroscience Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jungmin So
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young-Min Lim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kwang-Kuk Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Nam-Hee Kim
- Department of Neurology, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Tai-Seung Nam
- Department of Neurology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Sa-Yoon Kang
- Department of Neurology, Jeju National University School of Medicine, Jeju, Korea
| | - Jeeyoung Oh
- Department of Neurology, Konkuk University Medical Center, Seoul, Korea
| | - Seong-il Oh
- Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Eunhee Sohn
- Department of Neurology, Chungnam National University Hospital, Daejeon, Korea
| | - Ho Jin Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| |
Collapse
|
12
|
Quirant-Sánchez B, Mansilla MJ, Navarro-Barriuso J, Presas-Rodríguez S, Teniente-Serra A, Fondelli F, Ramo-Tello C, Martínez-Cáceres E. Combined Therapy of Vitamin D3-Tolerogenic Dendritic Cells and Interferon-β in a Preclinical Model of Multiple Sclerosis. Biomedicines 2021; 9:biomedicines9121758. [PMID: 34944573 PMCID: PMC8698295 DOI: 10.3390/biomedicines9121758] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/17/2022] Open
Abstract
Autologous antigen-specific therapies based on tolerogenic dendritic cells (tolDC) offer the possibility to treat autoimmune diseases by restoring homeostasis and targeting specifically autoreactive responses. Here, we explore the hypothesis that systemic inflammation occurring in autoimmune diseases, such as multiple sclerosis (MS), can generate a disease-specific environment able to alter the functionality of tolDC. In this context in fact, a combined therapy of tolDC with an immunomodulatory treatment could potentiate the beneficial effect of this antigen-specific cell therapy. For this purpose, we analyzed the efficacy of a combined therapy based on the use of vitamin D3 (VitD3)-tolDC plus interferon beta (IFN-beta) in MS. VitD3-tolDC were generated from healthy donors and MS patients and co-cultured with allogeneic peripheral blood mononuclear cells, in the presence or absence of IFN-beta. In vitro, VitD3-tolDC treatment reduced the percentage of activated T cells and allogeneic proliferation, whereas VitD3-tolDC+IFN-beta treatment enhanced the suppressive ability of VitD3-tolDC and, additionally, induced a shift towards a Th2 profile. To determine the clinical benefit of the combined therapy, C57BL/6-experimental autoimmune encephalomyelitis (EAE)-induced mice were treated with antigen-specific VitD3-tolDC and/or IFN-beta. Treatment of EAE mice with combined therapy ameliorated the disease course compared to each monotherapy. These results suggest that a combined therapy based on antigen-specific VitD3-tolDC and IFN-beta may represent a promising strategy for MS patients.
Collapse
Affiliation(s)
- Bibiana Quirant-Sánchez
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - María José Mansilla
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Juan Navarro-Barriuso
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Silvia Presas-Rodríguez
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain;
- Department of Medicine, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Aina Teniente-Serra
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Federico Fondelli
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Cristina Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain;
- Department of Medicine, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Correspondence: or (C.R.-T.); (E.M.-C.); Tel.: +34-93-497-8433 (C.R.-T.); +34-93-497-8666 (E.M.-C.)
| | - Eva Martínez-Cáceres
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Correspondence: or (C.R.-T.); (E.M.-C.); Tel.: +34-93-497-8433 (C.R.-T.); +34-93-497-8666 (E.M.-C.)
| |
Collapse
|
13
|
Multiple sclerosis patients have reduced resting and increased activated CD4 +CD25 +FOXP3 +T regulatory cells. Sci Rep 2021; 11:10476. [PMID: 34006899 PMCID: PMC8131694 DOI: 10.1038/s41598-021-88448-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/01/2021] [Indexed: 12/26/2022] Open
Abstract
Resting and activated subpopulations of CD4+CD25+CD127loT regulatory cells (Treg) and CD4+CD25+CD127+ effector T cells in MS patients and in healthy individuals were compared. Peripheral blood mononuclear cells isolated using Ficoll Hypaque were stained with monoclonal antibodies and analysed by flow cytometer. CD45RA and Foxp3 expression within CD4+ cells and in CD4+CD25+CD127loT cells identified Population I; CD45RA+Foxp3+, Population II; CD45RA−Foxp3hi and Population III; CD45RA−Foxp3+ cells. Effector CD4+CD127+ T cells were subdivided into Population IV; memory /effector CD45RA− CD25−Foxp3− and Population V; effector naïve CD45RA+CD25−Foxp3−CCR7+ and terminally differentiated RA+ (TEMRA) effector memory cells. Chemokine receptor staining identified CXCR3+Th1-like Treg, CCR6+Th17-like Treg and CCR7+ resting Treg. Resting Treg (Population I) were reduced in MS patients, both in untreated and treated MS compared to healthy donors. Activated/memory Treg (Population II) were significantly increased in MS patients compared to healthy donors. Activated effector CD4+ (Population IV) were increased and the naïve/ TEMRA CD4+ (Population V) were decreased in MS compared to HD. Expression of CCR7 was mainly in Population I, whereas expression of CCR6 and CXCR3 was greatest in Populations II and intermediate in Population III. In MS, CCR6+Treg were lower in Population III. This study found MS is associated with significant shifts in CD4+T cells subpopulations. MS patients had lower resting CD4+CD25+CD45RA+CCR7+ Treg than healthy donors while activated CD4+CD25hiCD45RA−Foxp3hiTreg were increased in MS patients even before treatment. Some MS patients had reduced CCR6+Th17-like Treg, which may contribute to the activity of MS.
Collapse
|
14
|
Malone K, Diaz Diaz AC, Shearer JA, Moore AC, Waeber C. The effect of fingolimod on regulatory T cells in a mouse model of brain ischaemia. J Neuroinflammation 2021; 18:37. [PMID: 33516262 PMCID: PMC7847573 DOI: 10.1186/s12974-021-02083-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/14/2021] [Indexed: 11/30/2022] Open
Abstract
Background The role of the immune system in stroke is well-recognised. Fingolimod, an immunomodulatory agent licensed for the management of relapsing-remitting multiple sclerosis, has been shown to provide benefit in rodent models of stroke. Its mechanism of action, however, remains unclear. We hypothesised fingolimod increases the number and/or function of regulatory T cells (Treg), a lymphocyte population which promotes stroke recovery. The primary aim of this study was to rigorously investigate the effect of fingolimod on Tregs in a mouse model of brain ischaemia. The effect of fingolimod in mice with common stroke-related comorbidities (ageing and hypercholesteremia) was also investigated. Methods Young (15–17 weeks), aged C57BL/6 mice (72–73 weeks), and ApoE−/− mice fed a high-fat diet (20–21 weeks) underwent permanent electrocoagulation of the left middle cerebral artery. Mice received either saline or fingolimod (0.5 mg/kg or 1 mg/kg) at 2, 24, and 48 h post-ischaemia via intraperitoneal injection. Another cohort of young mice (8–9, 17–19 weeks) received short-term (5 days) or long-term (10 days) fingolimod (0.5 mg/kg) treatment. Flow cytometry was used to quantify Tregs in blood, spleen, and lymph nodes. Immunohistochemistry was used to quantify FoxP3+ cell infiltration into the ischaemic brain. Results Fingolimod significantly increased the frequency of Tregs within the CD4+ T cell population in blood and spleen post-ischaemia in all three mouse cohorts compared to untreated ischemic mice. The highest splenic Treg frequency in fingolimod-treated mice was observed in ApoE−/− mice (9.32 ± 1.73% vs. 7.8 ± 3.01% in young, 6.09 ± 1.64% in aged mice). The highest circulating Treg frequency was also noted in ApoE−/− mice (8.39 ± 3.26% vs. 5.43 ± 2.74% in young, 4.56 ± 1.60% in aged mice). Fingolimod significantly increased the number of FoxP3+ cells in the infarct core of all mice. The most pronounced effects were seen when mice were treated for 10 days post-ischaemia. Conclusions Fingolimod increases Treg frequency in spleen and blood post-ischaemia and enhances the number of FoxP3+ cells in the ischaemic brain. The effect of fingolimod on this regulatory cell population may underlie its neuroprotective activity and could be exploited as part of future stroke therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02083-5.
Collapse
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Andrea C Diaz Diaz
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Jennifer A Shearer
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Anne C Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland. .,School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
15
|
Effect of Fingolimod and Interferon β-1α in Children With Multiple Sclerosis. Am J Ther 2020; 29:e99-e101. [PMID: 33021528 DOI: 10.1097/mjt.0000000000001276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Huber JE, Chang Y, Meinl I, Kümpfel T, Meinl E, Baumjohann D. Fingolimod Profoundly Reduces Frequencies and Alters Subset Composition of Circulating T Follicular Helper Cells in Multiple Sclerosis Patients. THE JOURNAL OF IMMUNOLOGY 2020; 204:1101-1110. [PMID: 32034063 DOI: 10.4049/jimmunol.1900955] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/12/2019] [Indexed: 12/22/2022]
Abstract
Fingolimod is an effective treatment for relapsing-remitting multiple sclerosis. It is well established that fingolimod, a modulator of the sphingosine-1-phosphate pathway, restrains the egress of CCR7+ lymphocytes from lymphatic tissues into the blood, thus resulting in reduced lymphocyte counts in peripheral blood. CXCR5+ T follicular helper (Tfh) cells provide help to B cells, are essential for the generation of potent Ab responses, and have been shown to be critically involved in the pathogenesis of several autoimmune diseases. Besides lymphoid tissue-resident Tfh cells, CXCR5+ circulating Tfh (cTfh) cells have been described in the blood, their numbers correlating with the magnitude of Tfh cells in lymphoid tissues. Although the effect of fingolimod on circulating lymphocyte subsets has been established, its effect on cTfh cells remains poorly understood. In this study, we found that although fingolimod strongly and disproportionally reduced cTfh cell frequencies, frequencies of activated cTfh cells were increased, and the composition of the cTfh cell pool was skewed toward a cTfh1 cell phenotype. The circulating T follicular regulatory cell subset and CXCR5+ CD8+ T cell frequencies were also strongly and disproportionally decreased after fingolimod treatment. In contrast, relative frequencies of CXCR5- memory Th cells as well as regulatory T and B cells were increased. In summary, these data provide new insights into fingolimod-induced compositional changes of lymphocyte populations in the blood, in particular cTfh cells, and thus contribute to a better understanding of the mechanism of action of fingolimod in multiple sclerosis patients.
Collapse
Affiliation(s)
- Johanna E Huber
- Institute for Immunology, Biomedical Center, Faculty of Medicine, LMU Munich, Planegg-Martinsried 82152, Germany; and
| | - Yinshui Chang
- Institute for Immunology, Biomedical Center, Faculty of Medicine, LMU Munich, Planegg-Martinsried 82152, Germany; and
| | - Ingrid Meinl
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Dirk Baumjohann
- Institute for Immunology, Biomedical Center, Faculty of Medicine, LMU Munich, Planegg-Martinsried 82152, Germany; and
| |
Collapse
|
17
|
Harris S, Tran JQ, Southworth H, Spencer CM, Cree BAC, Zamvil SS. Effect of the sphingosine-1-phosphate receptor modulator ozanimod on leukocyte subtypes in relapsing MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:7/5/e839. [PMID: 32737072 PMCID: PMC7413711 DOI: 10.1212/nxi.0000000000000839] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/17/2020] [Indexed: 12/28/2022]
Abstract
Objective To better understand ozanimod's mechanism of action (MOA), we conducted exploratory analyses from a phase 1 study to characterize ozanimod's effect on circulating leukocyte subsets in patients with relapsing multiple sclerosis. Methods An open-label pharmacodynamic study randomized patients to oral ozanimod hydrochloride (HCl) 0.5 (n = 13) or 1 mg/d (n = 11) for ∼12 weeks (including 7-day dose escalation). Circulating leukocyte subsets were quantified using flow cytometry (days 28, 56, and 85) and epigenetic cell counting (days 2, 5, 28, 56, and 85) and compared with baseline (day 1) using descriptive statistics. Results Ozanimod caused dose-dependent reductions in absolute lymphocyte counts. Observed by both methodologies, circulating CD19+ B- and CD3+ T-cell counts were reduced by >50% with ozanimod HCl 0.5 mg and >75% with 1 mg at day 85. Based on flow cytometry, ozanimod HCl 1 mg showed greater decreases in CD4+ than CD8+ T cells, greater decreases in both CD4+ and CD8+ central memory vs effector memory T cells, and reductions in mean CD4+ and CD8+ naive T cells by ≥90% at day 85. In the flow cytometry analysis, changes in monocytes, natural killer, and natural killer T cells were minimal. Using epigenetic cell counting, greater reductions for Th17 than T regulatory cells were determined. Conclusion Ozanimod induced dose-dependent reductions in circulating B- and T-cell counts and differential effects on naive and memory CD4+ and CD8+ T cells and CD19+ B cells. Data characterized with both a novel epigenetic cell-counting method and flow cytometry support ozanimod's MOA. Clinical trial registration: clinicaltrials.gov NCT02797015.
Collapse
Affiliation(s)
- Sarah Harris
- From Bristol-Myers Squibb Company (S.H., J.Q.T.), Princeton, NJ; Data Clarity Consulting Ltd. (H.S.), Stockport, England, United Kingdom; and Department of Neurology, Weill Institute for Neurosciences (C.M.S., B.A.C.C., S.S.Z.), and Program in Immunology (C.M.S., S.S.Z.), University of California San Francisco (UCSF).
| | - Jonathan Q Tran
- From Bristol-Myers Squibb Company (S.H., J.Q.T.), Princeton, NJ; Data Clarity Consulting Ltd. (H.S.), Stockport, England, United Kingdom; and Department of Neurology, Weill Institute for Neurosciences (C.M.S., B.A.C.C., S.S.Z.), and Program in Immunology (C.M.S., S.S.Z.), University of California San Francisco (UCSF)
| | - Harry Southworth
- From Bristol-Myers Squibb Company (S.H., J.Q.T.), Princeton, NJ; Data Clarity Consulting Ltd. (H.S.), Stockport, England, United Kingdom; and Department of Neurology, Weill Institute for Neurosciences (C.M.S., B.A.C.C., S.S.Z.), and Program in Immunology (C.M.S., S.S.Z.), University of California San Francisco (UCSF)
| | - Collin M Spencer
- From Bristol-Myers Squibb Company (S.H., J.Q.T.), Princeton, NJ; Data Clarity Consulting Ltd. (H.S.), Stockport, England, United Kingdom; and Department of Neurology, Weill Institute for Neurosciences (C.M.S., B.A.C.C., S.S.Z.), and Program in Immunology (C.M.S., S.S.Z.), University of California San Francisco (UCSF)
| | - Bruce A C Cree
- From Bristol-Myers Squibb Company (S.H., J.Q.T.), Princeton, NJ; Data Clarity Consulting Ltd. (H.S.), Stockport, England, United Kingdom; and Department of Neurology, Weill Institute for Neurosciences (C.M.S., B.A.C.C., S.S.Z.), and Program in Immunology (C.M.S., S.S.Z.), University of California San Francisco (UCSF)
| | - Scott S Zamvil
- From Bristol-Myers Squibb Company (S.H., J.Q.T.), Princeton, NJ; Data Clarity Consulting Ltd. (H.S.), Stockport, England, United Kingdom; and Department of Neurology, Weill Institute for Neurosciences (C.M.S., B.A.C.C., S.S.Z.), and Program in Immunology (C.M.S., S.S.Z.), University of California San Francisco (UCSF)
| |
Collapse
|
18
|
Boffa G, Bruschi N, Cellerino M, Lapucci C, Novi G, Sbragia E, Capello E, Uccelli A, Inglese M. Fingolimod and Dimethyl-Fumarate-Derived Lymphopenia is not Associated with Short-Term Treatment Response and Risk of Infections in a Real-Life MS Population. CNS Drugs 2020; 34:425-432. [PMID: 32193826 DOI: 10.1007/s40263-020-00714-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The association between treatment-related lymphopenia in multiple sclerosis, drug efficacy and the risk of infections is not yet fully understood. OBJECTIVE The objective of this study was to assess whether lymphopenia is associated with short-term treatment response and infection rate in a real-life multiple sclerosis population treated with fingolimod and dimethyl-fumarate. We assessed the associations between baseline absolute lymphocyte count and the lymphocyte mean percentage decrease at 6 and 12 months with treatment response and the occurrence of adverse events over 12 months in the entire cohort of patients and in the two treatment groups separately. METHODS This is a retrospective observational real-world study of patients with multiple sclerosis treated with fingolimod and dimethyl-fumarate at the MS Center of the University of Genoa between 2011 and 2018. Patients with at least 12 months of follow-up were eligible if [1] they had an Expanded Disability Status Scale assessment at baseline and 12 months after treatment onset, [2] they had undergone brain magnetic resonance imaging at baseline and after 12 months, and [3] absolute lymphocyte counts were available at baseline, 6 and 12 months. Patients shifting from dimethyl-fumarate to fingolimod or vice versa were excluded from the analysis. RESULTS In total, 137 and 75 patients treated with fingolimod and dimethyl-fumarate, respectively, were included in the analysis. At 12 months, fingolimod-treated patients were more likely to experience grade II and grade III lymphopenia compared with dimethyl-fumarate patients (p < 0.001, χ2 = 94) and had a higher lymphocyte mean percentage decrease (p < 0.001, U = 540). A higher number of previous therapies and a lower baseline absolute lymphocyte count were predictors of lymphopenia at 6 months (p = 0.047, odds ratio = 1.60 and p = 0.014, odds ratio = 1.1) and 12 months (p = 0.003, odds ratio = 1.97 and p = 0.023, odds ratio = 1.1). In fingolimod-treated patients only, female sex and a higher Expanded Disability Status Scale score were predictors of lymphopenia at 12 months (p = 0.006, odds ratio = 7.58 and p = 0.03, odds ratio = 1.56). Neither absolute lymphocyte count at 6 and 12 months nor the mean percentage decrease at 6 and 12 months predicted No Evidence of Disease Activity (NEDA-3) status at 1 year, the occurrence of relapses, disease activity on MRI or disability progression. CONCLUSIONS Our findings suggest that peripheral blood lymphocyte changes are not associated with short-term treatment response and with the rate of infections during fingolimod and dimethyl-fumarate treatment in real-world patients. Higher treatment exposure and a lower baseline absolute lymphocyte count are risk factors for lymphopenia development during fingolimod and dimethyl-fumarate therapy.
Collapse
Affiliation(s)
- Giacomo Boffa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Nicolò Bruschi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Maria Cellerino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Caterina Lapucci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Giovanni Novi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Elvira Sbragia
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Elisabetta Capello
- Ospedale Policlinico San Martino IRCCS, Largo Daneo 3, 16100, Genoa, Italy
| | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy.,Ospedale Policlinico San Martino IRCCS, Largo Daneo 3, 16100, Genoa, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy. .,Ospedale Policlinico San Martino IRCCS, Largo Daneo 3, 16100, Genoa, Italy.
| |
Collapse
|
19
|
Immunological Pattern in IgA Nephropathy. Int J Mol Sci 2020; 21:ijms21041389. [PMID: 32085673 PMCID: PMC7073027 DOI: 10.3390/ijms21041389] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022] Open
Abstract
The current gold-standard diagnostic technique for IgA nephropathy (IgAN), the leading form of primary glomerulonephritis, is renal biopsy. CD89 (the main IgA receptor) is expressed on the surface of monocytes and plays a role in disease pathogenesis. Immunocomplexes formed by sCD89 (soluble form) and Gd-IgA1 are related to disease prognosis. We hypothesize that reduced CD89 surface expression on monocytes may be a marker of disease severity. We aimed to analyze leukocyte subpopulations in peripheral blood and CD89 surface expression on monocytes in a prospective study of 22 patients and 12 healthy subjects (HS). Leukocyte subpopulations and CD89 expression were analyzed by flow cytometry. IgAN patients had a higher percentage of activated and effector memory CD4+ and CD8+ T lymphocytes, a lower percentage of transitional B lymphocytes and plasmablasts, and a higher percentage of CD56dimCD16+ NK cells and myeloid dendritic cells compared with HS. Correlations between reduced CD89 expression levels on nonclassical monocytes, histological findings of a poor prognosis on renal biopsy and baseline renal function were observed. IgAN patients show a characteristic immunological pattern in peripheral blood. A reduced expression level of CD89 on nonclassical monocytes identifies patients with a worse renal prognosis.
Collapse
|
20
|
Ghadiri M, Rezk A, Li R, Evans A, Giacomini PS, Barnett MH, Antel J, Bar-Or A. Pre-treatment T-cell subsets associate with fingolimod treatment responsiveness in multiple sclerosis. Sci Rep 2020; 10:356. [PMID: 31941953 PMCID: PMC6962338 DOI: 10.1038/s41598-019-57114-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 12/16/2019] [Indexed: 01/01/2023] Open
Abstract
Biomarkers predicting fingolimod (FTY) treatment response in relapsing-remitting multiple sclerosis (RRMS) are lacking. Here, we performed extensive functional immunophenotyping using multiparametric flow cytometry to examine peripheral immune changes under FTY treatment and explore biomarkers of FTY treatment response. From among 135 RRMS patients who initiated FTY in a 2-year multicentre observational study, 36 were classified as ‘Active’ or ‘Stable’ based on clinical and/or radiological activity on-treatment. Flow cytometric analysis of immune cell subsets was performed on pre- and on-treatment peripheral blood mononuclear cells (PBMC) samples. Decreased absolute counts of B cells and most T-cell subsets were seen on-treatment. Senescent CD8 + T cells, CD56 + T cells, CD56dim natural killer cells, monocytes and dendritic cells were not reduced in number and hence relatively increased in frequency on-treatment. An unbiased multiparametric and traditional manual analysis of T-cell subsets suggested a higher pre-treatment frequency of CD4 + central memory T cells (TCM) in patients who were subsequently Active versus Stable on-treatment. Lower pre-treatment terminally differentiated effector memory (TEMRA) cell frequencies were also seen in the subsequently Active cohort. Together, our data highlight differential effects of FTY on peripheral immune cell subsets and suggest that pre-treatment T-cell subset frequencies may have value in predicting FTY treatment response.
Collapse
Affiliation(s)
- Mahtab Ghadiri
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Ayman Rezk
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Center for Neuroinflammation and Experimental Therapeutics, and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Li
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Center for Neuroinflammation and Experimental Therapeutics, and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Paul S Giacomini
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Michael H Barnett
- Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Jack Antel
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Amit Bar-Or
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada. .,Center for Neuroinflammation and Experimental Therapeutics, and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Fingolimod reduces CXCR4-mediated B cell migration and induces regulatory B cells-mediated anti-inflammatory immune repertoire. Mult Scler Relat Disord 2019; 34:29-37. [DOI: 10.1016/j.msard.2019.06.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/24/2019] [Accepted: 06/16/2019] [Indexed: 12/21/2022]
|
22
|
Th1Th17 CM Lymphocyte Subpopulation as a Predictive Biomarker of Disease Activity in Multiple Sclerosis Patients under Dimethyl Fumarate or Fingolimod Treatment. Mediators Inflamm 2019; 2019:8147803. [PMID: 31346315 PMCID: PMC6617925 DOI: 10.1155/2019/8147803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/14/2019] [Accepted: 03/10/2019] [Indexed: 12/13/2022] Open
Abstract
Peripheral blood biomarkers able to predict disease activity in multiple sclerosis (MS) patients have not been identified yet. Here, we analyzed the immune phenotype of T lymphocyte subpopulations in peripheral blood samples from 66 RRMS patients under DMF (n = 22) or fingolimod (n = 44) treatment, by flow cytometry. A correlation study between the percentage and absolute cell number of each lymphocyte subpopulation with the presence of relapses or new MRI lesions during 12-month follow-up was performed. Patients who had undergone relapses showed at baseline higher percentage of Th1CM cells (relapsed: 11.60 ± 4.17%vs. nonrelapsed: 9.25 ± 3.17%, p < 0.05) and Th1Th17CM cells (relapsed: 15.65 ± 6.15%vs. nonrelapsed: 10.14 ± 4.05%, p < 0.01) before initiating DMF or fingolimod treatment. Kaplan-Meier analysis revealed that patients with Th1Th17CM (CD4+CCR7+CD45RA−CCR6+CXCR3+) cells > 11.48% had a 50% relapse-free survival compared to patients with Th1Th17CMcells < 11.48% whose relapse-free survival was 88% (p = 0.013, log-rank test). Additionally, a high percentage of Th1Th17CM cells was also found in patients with MRI activity (MRI activity: 14.02 ± 5.87%vs. no MRI activity: 9.82 ± 4.06%, p < 0.01). Our results suggest that the percentage of Th1Th17CM lymphocytes at baseline is a predictive biomarker of activity during the first 12 months of treatment, regardless of the treatment.
Collapse
|
23
|
Mansilla MJ, Navarro-Barriuso J, Presas-Rodríguez S, Teniente-Serra A, Quirant-Sánchez B, Ramo-Tello C, Martínez-Cáceres EM. Optimal response to dimethyl fumarate is mediated by a reduction of Th1-like Th17 cells after 3 months of treatment. CNS Neurosci Ther 2019; 25:995-1005. [PMID: 31066225 PMCID: PMC6698982 DOI: 10.1111/cns.13142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 04/01/2019] [Accepted: 04/07/2019] [Indexed: 01/08/2023] Open
Abstract
Aim Dimethyl fumarate (DMF) is one of the most promising therapies for relapsing‐remitting multiple sclerosis (RRMS) patients since it has shown immunomodulatory and neuroprotective effects. However, a percentage of RRMS patients do not exhibit an optimal response to DMF. The objective of this study was to identify early biomarkers of treatment response by analyzing changes in peripheral leukocyte subpopulations directly in whole blood samples. Methods A longitudinal and prospective study analyzing peripheral blood leukocyte subpopulations in 22 RRMS patients before initiating DMF treatment (baseline) and at 1, 3, 6, and 12 months of follow‐up was performed. Differences between no evidence of disease activity (NEDA) and ongoing disease activity (ODA) patients were analyzed. Results The beneficial effect of DMF was associated with a specific depletion of memory CD4+ and CD8+ T lymphocytes and B cells. Importantly, only NEDA patients showed (a) a shift from a pro‐ to an antiinflammatory profile, with an increase of Th2 cells and a decrease of Th1‐like Th17 lymphocytes; and (b) an increase of regulatory CD56bright NK cells. Conclusion The optimal response to DMF is mediated by a shift to antiinflammatory and immunoregulatory profile, which puts forward Th1‐like Th17 lymphocytes as a potential early biomarker of treatment response.
Collapse
Affiliation(s)
- María José Mansilla
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Juan Navarro-Barriuso
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Silvia Presas-Rodríguez
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Badalona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Aina Teniente-Serra
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Bibiana Quirant-Sánchez
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Cristina Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Badalona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Eva María Martínez-Cáceres
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
24
|
Quirant‐Sánchez B, Hervás‐García JV, Teniente‐Serra A, Brieva L, Moral‐Torres E, Cano A, Munteis E, Mansilla MJ, Presas‐Rodriguez S, Navarro‐Barriuso J, Ramo‐Tello C, Martínez‐Cáceres EM. Predicting therapeutic response to fingolimod treatment in multiple sclerosis patients. CNS Neurosci Ther 2018; 24:1175-1184. [PMID: 29656444 PMCID: PMC6489963 DOI: 10.1111/cns.12851] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 03/01/2018] [Accepted: 03/05/2018] [Indexed: 12/20/2022] Open
Abstract
AIMS Fingolimod, an orally active immunomodulatory drug for relapsing-remitting multiple sclerosis (RRMS), sequesters T cells in lymph nodes through functional antagonism of the sphingosine-1-phosphate receptor, reducing the number of potential autoreactive cells that migrate to the central nervous system. However, not all RRMS patients respond to this therapy. Our aim was to test the hypothesis that by immune-monitoring RRMS patient's leukocyte subpopulations it is possible to find biomarkers associated with clinical response to fingolimod. METHODS Prospective study. Analysis of peripheral blood mononuclear cell subpopulations by multiparametric flow cytometry, at baseline and +1, +3, +6, +12 months of follow-up in 40 RRMS patients starting fingolimod therapy. RESULTS Fingolimod treatment induced a severe lymphopenia affecting mainly T and B cells. A relative increase in Treg (memory Treg : 3.8 ± 1.0% baseline vs 8.8 ± 4.4% month +1; activated Treg : 1.5 ± 0.7% baseline vs 3.7 ± 2.1% month +1, P < 0.001) as well as transitional B cells (10.5 ± 12.3% baseline vs 18.7 ± 14.6% month +1, P < 0.001) was observed. Interestingly, lymphocyte subpopulations were already at baseline significantly different in responder patients. The percentage of recent thymic emigrants (RTE) used to stratify fingolimod responder, and no responder patients was the best biomarker (4.0 ± 1.4% vs 7.4 ± 1.9%, respectively [P < 0.001]). CONCLUSION The results support that immune-monitoring of lymphocyte subpopulations in peripheral blood is a promising tool to select RRMS candidate for fingolimod treatment.
Collapse
Affiliation(s)
- Bibiana Quirant‐Sánchez
- Immunology DivisionGermans Trias i Pujol University Hospital and Research InstituteBarcelonaSpain
- Department of Cell Biology, Physiology and ImmunologyUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - José V. Hervás‐García
- Multiple Sclerosis UnitDepartment of NeurosciencesGermans Trias i Pujol University HospitalBarcelonaSpain
| | - Aina Teniente‐Serra
- Immunology DivisionGermans Trias i Pujol University Hospital and Research InstituteBarcelonaSpain
- Department of Cell Biology, Physiology and ImmunologyUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Luis Brieva
- Neurology Department of Hospital Arnau VilanovaLeridaSpain
| | - Ester Moral‐Torres
- Neurology Department of Hospital San Joan Despi Moises BroggiBarcelonaSpain
| | - Antonio Cano
- Neurology Department of Hospital de MataróBarcelonaSpain
| | | | - María J. Mansilla
- Immunology DivisionGermans Trias i Pujol University Hospital and Research InstituteBarcelonaSpain
- Department of Cell Biology, Physiology and ImmunologyUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Silvia Presas‐Rodriguez
- Multiple Sclerosis UnitDepartment of NeurosciencesGermans Trias i Pujol University HospitalBarcelonaSpain
| | - Juan Navarro‐Barriuso
- Immunology DivisionGermans Trias i Pujol University Hospital and Research InstituteBarcelonaSpain
- Department of Cell Biology, Physiology and ImmunologyUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Cristina Ramo‐Tello
- Multiple Sclerosis UnitDepartment of NeurosciencesGermans Trias i Pujol University HospitalBarcelonaSpain
| | - Eva M. Martínez‐Cáceres
- Immunology DivisionGermans Trias i Pujol University Hospital and Research InstituteBarcelonaSpain
- Department of Cell Biology, Physiology and ImmunologyUniversitat Autònoma de BarcelonaBarcelonaSpain
| |
Collapse
|
25
|
Kaufmann M, Haase R, Proschmann U, Ziemssen T, Akgün K. Real World Lab Data: Patterns of Lymphocyte Counts in Fingolimod Treated Patients. Front Immunol 2018; 9:2669. [PMID: 30524432 PMCID: PMC6256977 DOI: 10.3389/fimmu.2018.02669] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
Objective: Fingolimod is approved for the treatment of highly active relapsing remitting multiple sclerosis (MS) patients and acts by its unique mechanism of action via sphingosine-1-phosphate receptor-modulation. Although fingolimod-associated lymphopenia is a well-known phenomenon, the exact cause for the intra- and inter-individual differences of the fluctuation of lymphocyte count and its subtypes is still subject of debate. In this analysis, we aim to estimate the significance of the individual variation of distinct lymphocyte subsets for differences in absolute lymphocyte decrease in fingolimod treated patients and discuss how different lymphocyte subset patterns are related to clinical presentation in a long-term real life setting. Methods/Design: One hundred and thirteen patients with MS were characterized by complete blood cell count and immune cell phentopying of peripheral lymphocyte subsets before, at month 1 and every 3 months up to 36 months of fingolimod treatment. In addition, patients were monitored regarding clinical parameters (relapses, disability, MRI). Results: There was no significant association of baseline lymphocyte count and lymphocyte subtypes with lymphocyte decrease after fingolimod start. The initial drop of the absolute lymphocyte count could not predict the level of lymphocyte count during steady state on fingolimod. Variable CD8+ T cell and NK cell counts account for the remarkable intra- and inter-individual differences regarding initial drop and steady state level of lymphocyte count during fingolimod treatment, whereas CD4+ T cells and B cells mostly present a quite uniform decrease in all treated patients. Selected patients with lymphocyte count >1.0 GPT/l differed by higher CD8+ T cells and NK cell counts compared to lymphopenic patients but presented comparable clinical effectiveness during treatment. Conclusion: Monitoring of the absolute lymphocyte count at steady state seems to be a rough estimate of fingolimod induced lymphocyte redistribution. Our results suggest, that evaluation of distinct lymphocyte subsets as CD4+ T cells allow a more detailed evaluation to weigh and interpret degree of lymphopenia and treatment response in fingolimod treated patients.
Collapse
Affiliation(s)
- Maxi Kaufmann
- MS Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| | - Rocco Haase
- MS Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| | - Undine Proschmann
- MS Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| | - Tjalf Ziemssen
- MS Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| | - Katja Akgün
- MS Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
| |
Collapse
|
26
|
Moreno-Torres I, González-García C, Marconi M, García-Grande A, Rodríguez-Esparragoza L, Elvira V, Ramil E, Campos-Ruíz L, García-Hernández R, Al-Shahrour F, Fustero-Torre C, Sánchez-Sanz A, García-Merino A, Sánchez López AJ. Immunophenotype and Transcriptome Profile of Patients With Multiple Sclerosis Treated With Fingolimod: Setting Up a Model for Prediction of Response in a 2-Year Translational Study. Front Immunol 2018; 9:1693. [PMID: 30090102 PMCID: PMC6068231 DOI: 10.3389/fimmu.2018.01693] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/10/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Fingolimod is a functional sphingosine-1-phosphate antagonist approved for the treatment of multiple sclerosis (MS). Fingolimod affects lymphocyte subpopulations and regulates gene expression in the lymphocyte transcriptome. Translational studies are necessary to identify cellular and molecular biomarkers that might be used to predict the clinical response to the drug. In MS patients, we aimed to clarify the differential effects of fingolimod on T, B, and natural killer (NK) cell subsets and to identify differentially expressed genes in responders and non-responders (NRs) to treatment. MATERIALS AND METHODS Samples were obtained from relapsing-remitting multiple sclerosis patients before and 6 months after starting fingolimod. Forty-eight lymphocyte subpopulations were measured by flow cytometry based on surface and intracellular marker analysis. Transcriptome sequencing by next-generation technologies was used to define the gene expression profiling in lymphocytes at the same time points. NEDA-3 (no evidence of disease activity) and NEDA-4 scores were measured for all patients at 1 and 2 years after beginning fingolimod treatment to investigate an association with cellular and molecular characteristics. RESULTS Fingolimod affects practically all lymphocyte subpopulations and exerts a strong effect on genetic transcription switching toward an anti-inflammatory and antioxidant response. Fingolimod induces a differential effect in lymphocyte subpopulations after 6 months of treatment in responder and NR patients. Patients who achieved a good response to the drug compared to NR patients exhibited higher percentages of NK bright cells and plasmablasts, higher levels of FOXP3, glucose phosphate isomerase, lower levels of FCRL1, and lower Expanded Disability Status Scale at baseline. The combination of these possible markers enabled us to build a probabilistic linear model to predict the clinical response to fingolimod. CONCLUSION MS patients responsive to fingolimod exhibit a recognizable distribution of lymphocyte subpopulations and a different pretreatment gene expression signature that might be useful as a biomarker.
Collapse
Affiliation(s)
- Irene Moreno-Torres
- Neuroimmunology Unit, Puerta de Hierro-Segovia de Arana Health Research Institute, Madrid, Spain
- Autonomous University of Madrid, Madrid, Spain
| | - Coral González-García
- Neuroimmunology Unit, Puerta de Hierro-Segovia de Arana Health Research Institute, Madrid, Spain
| | - Marco Marconi
- Centre for Plant Biotechnology and Genomics, Madrid, Spain
| | - Aranzazu García-Grande
- Flow Cytometry Core Facility, Puerta de Hierro-Segovia de Arana Health Research Institute, Madrid, Spain
| | | | - Víctor Elvira
- IMT Lille Douai & CRIStAL, Univ. de Lille, Douai, France
| | - Elvira Ramil
- Sequencing Core Facility, Puerta de Hierro-Segovia de Arana Health Research Institute, Madrid, Spain
| | - Lucía Campos-Ruíz
- Neuroimmunology Unit, Puerta de Hierro-Segovia de Arana Health Research Institute, Madrid, Spain
| | - Ruth García-Hernández
- Neuroimmunology Unit, Puerta de Hierro-Segovia de Arana Health Research Institute, Madrid, Spain
- Autonomous University of Madrid, Madrid, Spain
| | - Fátima Al-Shahrour
- Bioinformatics Unit of Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Coral Fustero-Torre
- Bioinformatics Unit of Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Alicia Sánchez-Sanz
- Neuroimmunology Unit, Puerta de Hierro-Segovia de Arana Health Research Institute, Madrid, Spain
| | - Antonio García-Merino
- Neuroimmunology Unit, Puerta de Hierro-Segovia de Arana Health Research Institute, Madrid, Spain
- Autonomous University of Madrid, Madrid, Spain
- Neurology Department, Puerta de Hierro University Hospital, Madrid, Spain
- Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain
| | - Antonio José Sánchez López
- Neuroimmunology Unit, Puerta de Hierro-Segovia de Arana Health Research Institute, Madrid, Spain
- Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain
- Biobank, Puerta de Hierro University Hospital-IDIPHISA, Madrid, Spain
| |
Collapse
|