1
|
Harris S, Feagan BG, Hanauer S, Vermeire S, Ghosh S, Yan J, Wu C, Hu Y, Maddux R, Wolf DC, D'Haens G. Ozanimod Differentially Impacts Circulating Lymphocyte Subsets in Patients with Moderately to Severely Active Crohn's Disease. Dig Dis Sci 2024; 69:2044-2054. [PMID: 38568396 PMCID: PMC11162376 DOI: 10.1007/s10620-024-08391-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/13/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Ozanimod showed efficacy and safety in the phase 2 STEPSTONE study conducted in patients with moderately to severely active Crohn's disease. AIMS This analysis assessed the effects of ozanimod on circulating lymphocytes in Crohn's disease. METHODS Patients received ozanimod 0.92 mg for 12 weeks. Lymphocyte subtypes were evaluated using multicolor flow analysis on blood samples collected before treatment and on Week 12. Absolute lymphocyte count changes were analyzed by Wilcoxon signed rank tests. Disease activity changes and efficacy outcomes were evaluated at Week 12, and associations with lymphocyte subtype levels were assessed using Spearman's correlation and logistic regression. RESULTS Reductions in median total T, Th, and cytotoxic T cells occurred at Week 12 (45.4%-76.8%), with reductions in most subtypes of 47.5% to 91.3% (P < 0.001). CD8+ terminally differentiated effector memory cells were largely unaffected (median change, - 19%; P = 0.44). Reductions in median total B cells occurred at Week 12 (76.7%), with reductions in subtypes of 71.4% to 81.7% (P < 0.001). Natural killer and monocyte cell counts were unchanged. Greater baseline levels and changes in nonswitched memory B cells were significantly associated with clinical, endoscopic, and histologic efficacy (P < 0.05, all comparisons). CONCLUSIONS Ozanimod reduced circulating levels of all B-cell and most T-cell subsets but not monocytes or natural killer cells. Key subsets relevant to immune surveillance were not reduced, supporting the low risk of infection and malignancy with ozanimod in chronic inflammatory diseases. Levels of nonswitched memory B cells were associated with efficacy, providing a potential marker for ozanimod response. TRIAL REGISTRATION ClinicalTrials.gov: NCT02531113, EudraCT: 2015-002025-19.
Collapse
Affiliation(s)
- Sarah Harris
- Bristol Myers Squibb, Princeton, NJ, USA.
- Bristol Myers Squibb, 3033 Science Park Rd, San Diego, CA, 92121, USA.
| | - Brian G Feagan
- Robarts Clinical Trials, Robarts Research Institute, Western University, London, ON, Canada
| | - Stephen Hanauer
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Subrata Ghosh
- College of Medicine and Health, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jim Yan
- Laboratory Corporation of America, Durham, NC, USA
| | - Chun Wu
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Yanhua Hu
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | - Douglas C Wolf
- Center for Crohn's Disease & Ulcerative Colitis, Atlanta Gastroenterology Associates, Atlanta, GA, USA
| | - Geert D'Haens
- Inflammatory Bowel Disease Center, Amsterdam University Medical Center, Amsterdam, Netherlands
| |
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
|
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
|
4
|
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
|
5
|
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
|
6
|
Teniente-Serra A, Pizarro E, Quirant-Sánchez B, Fernández MA, Vives-Pi M, Martinez-Caceres EM. Identifying Changes in Peripheral Lymphocyte Subpopulations in Adult Onset Type 1 Diabetes. Front Immunol 2021; 12:784110. [PMID: 34938295 PMCID: PMC8685245 DOI: 10.3389/fimmu.2021.784110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/18/2021] [Indexed: 01/11/2023] Open
Abstract
T- and B-lymphocytes play an important role in the pathogenesis of type 1 diabetes (T1D), a chronic disease caused by the autoimmune destruction of the insulin-producing cells in the pancreatic islets. Flow cytometry allows their characterization in peripheral blood, letting to investigate changes in cellular subpopulations that can provide insights in T1D pathophysiology. With this purpose, CD4+ and CD8+ T cells (including naïve, central memory, effector memory and terminally differentiated effector (TEMRA), Th17 and Tregs) and B cells subsets (naïve, unswitched memory, switched memory and transitional B cells) were analysed in peripheral blood of adult T1D patients at disease onset and after ≥2 years using multiparametric flow cytometry. Here we report changes in the percentage of early and late effector memory CD4+ and CD8+ T cells as well as of naïve subsets, regulatory T cells and transitional B cells in peripheral blood of adult patients at onset of T1D when compared with HD. After 2 years follow-up these changes were maintained. Also, we found a decrease in percentage of Th17 and numbers of T cells with baseline. In order to identify potential biomarkers of disease, ROC curves were performed being late EM CD4 T cell subset the most promising candidate. In conclusion, the observed changes in the percentage and/or absolute number of lymphocyte subpopulations of adult T1D patients support the hypothesis that effector cells migrate to the pancreas and this autoimmune process perseveres along the disease. Moreover, multiparametric flow allows to identify those subsets with potential to be considered biomarkers of disease.
Collapse
Affiliation(s)
- Aina Teniente-Serra
- Immunology Division, Clinical Laboratory MetroNord (LCMN), Germans Trias i Pujol University Hospital and Research Institute (IGTP), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Spain
| | - Eduarda Pizarro
- Endocrinology Department, Hospital de Mataró, Barcelona, Spain
| | - Bibiana Quirant-Sánchez
- Immunology Division, Clinical Laboratory MetroNord (LCMN), Germans Trias i Pujol University Hospital and Research Institute (IGTP), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Spain
| | - Marco A Fernández
- Flow Cytometry Facility, Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Marta Vives-Pi
- Immunology Division, Clinical Laboratory MetroNord (LCMN), Germans Trias i Pujol University Hospital and Research Institute (IGTP), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Spain
| | - Eva M Martinez-Caceres
- Immunology Division, Clinical Laboratory MetroNord (LCMN), Germans Trias i Pujol University Hospital and Research Institute (IGTP), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Spain
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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]
|
10
|
Teniente-Serra A, Soldevila B, Quirant-Sánchez B, Fernández MA, Ester Condins A, Puig-Domingo M, Pujol-Borrell R, Martínez-Cáceres EM. Distinct pattern of peripheral lymphocyte subsets in Graves' disease with persistency of anti-TSHR autoantibodies. Autoimmunity 2019; 52:220-227. [PMID: 31366254 DOI: 10.1080/08916934.2019.1646253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background: Graves' disease (GD) is characterized by the production of autoantibodies against the TSHR (TRAbs). With long-term treatment, serum concentrations of TRAbs decline but in some patients, despite being clinically stable, TRAbs persist for many years.Objective: To investigate whether GD patients with persistence of TRAbs constitute a subset of patients that could be identified by phenotypic analysis of circulating lymphocytes, suggesting disease heterogeneity.Materials and methods: Peripheral blood lymphocytes (including naïve, memory and effector T and B cells, Th17, regulatory T cells (Treg), recent thymic emigrants (RTEs) and double positive CD4+CD8+ (DP) cells) were analysed by flow cytometry in a cross-sectional study in 25 clinically stable GD patients, five patients at onset of GD disease and 40 healthy donors (HDs).Results: GD patients with persistence of TRAbs showed a lower percentage of Treg and lower absolute numbers of central and effector memory CD8+ T cells than HD. No differences in RTEs were found in peripheral blood from GD patients compared to HD. Stable GD patients had higher percentage of DP cells of effector phenotype than HD.Conclusions: Using extensive phenotypic analysis of lymphocyte subpopulations, it is possible to detect changes that help to identify patients with persistent TSHR antibodies and may contribute to understand why the autoimmune response is maintained.
Collapse
Affiliation(s)
- Aina Teniente-Serra
- Immunology Division, LCMN Germans Trias i Pujol University Hospital Badalona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
| | - Berta Soldevila
- Endocrinology and Nutrition Department, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Bibiana Quirant-Sánchez
- Immunology Division, LCMN Germans Trias i Pujol University Hospital Badalona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
| | - Marco A Fernández
- Flow Cytometry Facility, Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | | | - Manuel Puig-Domingo
- Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain.,Endocrinology and Nutrition Department, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Ricardo Pujol-Borrell
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Immunology Division, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Eva M Martínez-Cáceres
- Immunology Division, LCMN Germans Trias i Pujol University Hospital Badalona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
Rasche L, Paul F. Ozanimod for the treatment of relapsing remitting multiple sclerosis. Expert Opin Pharmacother 2018; 19:2073-2086. [PMID: 30407868 DOI: 10.1080/14656566.2018.1540592] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Ozanimod is a selective sphingosine 1-phosphate receptor 1 and 5 modulator under development by Celgene, for the treatment of relapsing remitting multiple sclerosis. Extensive clinical experience has become available for the related compound fingolimod, favoring the sphingosine 1-phosphate therapeutic concept. Off-target effects have been attributed to its low receptor specificity and have prompted the development of next generation sphingosine 1-phosphate receptor modulators. Areas covered: The authors evaluate the literature of ozanimod, using the PubMed database as well as repositories of the European Committee for Treatment and Research in Multiple Sclerosis and the American and European Academy of Neurology. Specifically, the authors cover and discuss the preclinical data on ozanimod, pharmacokinetics and dynamics, and data on efficacy and safety from the pivotal trials. Expert opinion: Superiority of ozanimod over intramuscular interferon β-1a with regard to reduction in annualized relapse rate and magnetic resonance imaging outcomes has been shown in two phase III trials. The beneficial effect on brain volume and gray matter loss are encouraging and in line with data on other newer immunomodulators. Ozanimod is a valuable contribution to the therapeutic armamentarium in MS, although the effect on disability progression is unclear and requires further investigations.
Collapse
Affiliation(s)
- Ludwig Rasche
- a NeuroCure Clinical Research Center , Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health , Berlin , Germany
| | - Friedemann Paul
- a NeuroCure Clinical Research Center , Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health , Berlin , Germany.,b Experimental and Clinical Research Center , Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health , Berlin , Germany.,c Department of Neurology , Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health , Berlin , Germany
| |
Collapse
|
16
|
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
|
17
|
Chen C, Zhou Y, Wang J, Yan Y, Peng L, Qiu W. Dysregulated MicroRNA Involvement in Multiple Sclerosis by Induction of T Helper 17 Cell Differentiation. Front Immunol 2018; 9:1256. [PMID: 29915595 PMCID: PMC5994557 DOI: 10.3389/fimmu.2018.01256] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/18/2018] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system. Growing evidence has proven that T helper 17 (Th17) cells are one of the regulators of neuroinflammation mechanisms in MS disease. Researchers have demonstrated that some microRNAs (miRNAs) are associated with disease activity and duration, even with different MS patterns. miRNAs regulate CD4+ T cells to differentiate toward various T cell subtypes including Th17 cells. In this review, we discuss the possible mechanisms of miRNAs in MS pathophysiology by regulating CD4+ T cell differentiation into Th17 cells, and potential miRNA targets for current disease-modifying treatments.
Collapse
Affiliation(s)
- Chen Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yifan Zhou
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingqi Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yaping Yan
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Lisheng Peng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Akgün K, Kaufmann M, Ziemssen T. Comment on Y.D. Fragoso et al.: “Lymphocyte count in peripheral blood is not associated with the level of clinical response to treatment with fingolimod” Multiple Sclerosis and Related Disorders 2017. Mult Scler Relat Disord 2018; 22:68-69. [DOI: 10.1016/j.msard.2018.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/15/2018] [Indexed: 10/17/2022]
|
19
|
Angerer IC, Hecker M, Koczan D, Roch L, Friess J, Rüge A, Fitzner B, Boxberger N, Schröder I, Flechtner K, Thiesen HJ, Winkelmann A, Meister S, Zettl UK. Transcriptome profiling of peripheral blood immune cell populations in multiple sclerosis patients before and during treatment with a sphingosine-1-phosphate receptor modulator. CNS Neurosci Ther 2018; 24:193-201. [PMID: 29314605 DOI: 10.1111/cns.12793] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022] Open
Abstract
AIMS Fingolimod is a sphingosine-1-phosphate (S1P) receptor modulator approved for the treatment of the relapsing form of multiple sclerosis (MS). It prevents the egress of lymphocyte subpopulations from lymphoid tissues into the circulation. Here, we explored the broad effects of fingolimod on gene expression in different immune cell subsets. METHODS Utilizing 150 high-resolution microarrays from Affymetrix, we obtained the transcriptome profiles of 5 cell populations, which were separated from the peripheral blood of MS patients prior to and following oral administration of fingolimod. RESULTS After 3 months of treatment, significant transcriptome shifts were seen in CD4+ and CD8+ cells, which is mainly attributable to the selective homing of naive T cells and central memory T cells. Although the number of B cells was greatly reduced in the blood of fingolimod-treated MS patients, the analysis of differential expression in CD19+ cells identified only a small set of 42 genes, which indicated a slightly higher frequency of transitional B cells. The transcriptome signatures of CD14+ monocytes and CD56+ natural killer cells were not affected. CONCLUSION Our study corroborates changes in the composition of circulating immune cells in response to fingolimod and delineates the respective implications at the RNA level. Our data may be valuable for comparing the effects of novel S1P receptor modulating agents, which may be a therapeutic option for patients with secondary progressive MS as well.
Collapse
Affiliation(s)
- Ines C Angerer
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany
| | - Michael Hecker
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany.,Steinbeis Transfer Center for Proteome Analysis, Rostock, Germany
| | - Dirk Koczan
- Steinbeis Transfer Center for Proteome Analysis, Rostock, Germany.,Institute of Immunology, University of Rostock, Rostock, Germany
| | - Luisa Roch
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany
| | - Jörg Friess
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany
| | - Annelen Rüge
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany
| | - Brit Fitzner
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany.,Steinbeis Transfer Center for Proteome Analysis, Rostock, Germany
| | - Nina Boxberger
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany
| | - Ina Schröder
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany
| | | | - Hans-Jürgen Thiesen
- Steinbeis Transfer Center for Proteome Analysis, Rostock, Germany.,Institute of Immunology, University of Rostock, Rostock, Germany
| | - Alexander Winkelmann
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany
| | - Stefanie Meister
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany
| | - Uwe K Zettl
- Department of Neurology, Division of Neuroimmunology, University of Rostock, Rostock, Germany
| |
Collapse
|
20
|
Lymphocyte count in peripheral blood is not associated with the level of clinical response to treatment with fingolimod. Mult Scler Relat Disord 2018; 19:105-108. [DOI: 10.1016/j.msard.2017.11.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 11/20/2022]
|
21
|
Role of Immunological Memory Cells as a Therapeutic Target in Multiple Sclerosis. Brain Sci 2017; 7:brainsci7110148. [PMID: 29112130 PMCID: PMC5704155 DOI: 10.3390/brainsci7110148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 12/14/2022] Open
Abstract
Pharmacological targeting of memory cells is an attractive treatment strategy in various autoimmune diseases, such as psoriasis and rheumatoid arthritis. Multiple sclerosis is the most common inflammatory disorder of the central nervous system, characterized by focal immune cell infiltration, activation of microglia and astrocytes, along with progressive damage to myelin sheaths, axons, and neurons. The current review begins with the identification of memory cell types in the previous literature and a recent description of the modulation of these cell types in T, B, and resident memory cells in the presence of different clinically approved multiple sclerosis drugs. Overall, this review paper tries to determine the potential of memory cells to act as a target for the current or newly-developed drugs.
Collapse
|
22
|
Thomas K, Proschmann U, Ziemssen T. Fingolimod hydrochloride for the treatment of relapsing remitting multiple sclerosis. Expert Opin Pharmacother 2017; 18:1649-1660. [PMID: 28844164 DOI: 10.1080/14656566.2017.1373093] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Fingolimod was the first oral and the first in class disease modifying treatment in multiple sclerosis that acts as sphingosine-1-phospathe receptor agonist. Since approval in 2010 there is a growing experience with fingolimod use in clinical practice, but also next-generation sphingosin-1-receptor agonists in ongoing clinical trials. Growing evidence demonstrates additional effects beyond impact on lymphocyte circulation, highlighting further promising targets in multiple sclerosis therapy. Areas covered: Here we present a systematic review using PubMed database searching and expert opinion on fingolimod use in clinical practice. Long-term data of initial clinical trials and post-marketing evaluations including long-term efficacy, safety, tolerability and management especially within growing disease modifying treatment options and pre-treatment constellation in multiple sclerosis patients are critically discussed. Furthermore novel findings in mechanism of actions and prospective on additional use in progressive forms in multiple sclerosis are presented. Expert opinion: There is an extensive long-term experience on fingolimod use in clinical practice demonstrating the favorable benefit-risk of this drug. Using a defined risk management approach experienced MS clinicians should apply fingolimod after critical choice of patients and review of clinical aspects. Further studies are essential to discuss additional benefit in progressive forms in multiple sclerosis.
Collapse
Affiliation(s)
- Katja Thomas
- a Center of Clinical Neuroscience , University Hospital, Dresden , Dresden , Germany
| | - Undine Proschmann
- a Center of Clinical Neuroscience , University Hospital, Dresden , Dresden , Germany
| | - Tjalf Ziemssen
- a Center of Clinical Neuroscience , University Hospital, Dresden , Dresden , Germany
| |
Collapse
|
23
|
Fingolimod alters the transcriptome profile of circulating CD4+ cells in multiple sclerosis. Sci Rep 2017; 7:42087. [PMID: 28155899 PMCID: PMC5290459 DOI: 10.1038/srep42087] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/04/2017] [Indexed: 01/09/2023] Open
Abstract
Multiple sclerosis is a demyelinating disease affecting the central nervous system. T cells are known to contribute to this immune-mediated condition. Fingolimod modulates sphingosine-1-phosphate receptors, thereby preventing the egress of lymphocytes, especially CCR7-expressing CD8+ and CD4+ T cells, from lymphoid tissues. Using Affymetrix Human Transcriptome Arrays (HTA 2.0), we performed a transcriptome profiling analysis of CD4+ cells obtained from the peripheral blood of patients with highly active relapsing-remitting multiple sclerosis. The samples were drawn before the first administration of fingolimod as well as 24 hours and 3 months after the start of therapy. Three months after treatment initiation, 890 genes were found to be differentially expressed with fold-change >2.0 and t-test p-value < 0.001, among them several microRNA precursors. A subset of 272 genes were expressed at lower levels, including CCR7 as expected, while 618 genes showed an increase in expression, e.g., CCR2, CX3CR1, CD39, CD58 as well as LYN, PAK1 and TLR2. To conclude, we studied the gene expression of CD4+ cells to evaluate the effects of fingolimod treatment, and we identified 890 genes to be altered in expression after continuous drug administration. T helper cells circulating in the blood during fingolimod therapy present a distinct gene expression signature.
Collapse
|