1
|
Guerra T, Caputo F, Bianco A, Paolicelli D, Iaffaldano P. Long-Term Evaluation of Effectiveness and Immunological Implications of Ocrelizumab in a Real-World Cohort. Drugs Real World Outcomes 2025:10.1007/s40801-025-00486-x. [PMID: 40085374 DOI: 10.1007/s40801-025-00486-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Extended follow-up data from real-world cohorts of patients with multiple sclerosis treated with ocrelizumab (OCR) are becoming widely available. This monocentric retrospective study aimed to evaluate the long-term effectiveness of OCR and its impact on immunoglobulin (Ig) levels, lymphocyte subsets, and infections in a cohort of patients with relapsing remitting, primary progressive, and secondary progressive multiple sclerosis. METHODS Patients followed at the Multiple Sclerosis Center of Bari with ≥ 2 years of OCR treatment were retrospectively recruited in 2024. Twelve-month confirmed disability worsening, improvement, and the annualized relapse rate before and after treatment start were estimated and follow-up magnetic resonance imaging scans were collected. Changes in IgG/IgM/IgA levels from baseline for up to 6 years of OCR treatment and serum levels of T CD4+, T CD8+, and natural killer lymphocytes were assessed. Infection occurrence, type, and outcomes were investigated. Multivariable Cox models examined the association of clinical and radiological baseline factors with the risk of confirmed disability worsening and the relationship of infections with clinical-laboratoristic risk factors. RESULTS The final cohort retrieved 140 patients (80 relapsing remitting, 37 primary progressive, 23 secondary progressive) with a median (interquartile range) follow-up after treatment start of 4.62 (4.10-5.04) years. In the entire cohort, the mean annualized relapse rate decreased from 0.61 in the year before the start of OCR treatment to 0.02 in the second year, thereafter all patients in our cohort remained free of relapses and magnetic resonance imaging activity. The overall percentage of stable patients increased from the second to the fourth year, in parallel with a reduction in patients with confirmed disability worsening. A multifocal onset and the presence of at least two relapses before the start of OCR treatment were significant (p < 0.05) risk factors of confirmed disability worsening. During the follow-up, a reduction in the IgG serum level was observed, which further decreased, becoming stable after the third year. Immunoglobulin M levels slightly decreased over time, whereas IgA levels remained stable. No changes for T CD4+, natural killer cell absolute number, and a slight reduction in T CD8+ lymphocytes during follow-up were observed. Ocrelizumab did not determine a significant infection risk in the long term and no association was observed with Ig levels and severe infections. CONCLUSIONS Ocrelizumab prevented disease activity over the long term and its effect on the immune system did not determine a significant infection risk in most patients.
Collapse
Affiliation(s)
- Tommaso Guerra
- Department of Translational Biomedicine and Neurosciences (DiBraiN), Multiple Sclerosis Center - Policlinico of Bari, University of Bari "Aldo Moro", Bari, Italy
| | - Francesca Caputo
- Department of Translational Biomedicine and Neurosciences (DiBraiN), Multiple Sclerosis Center - Policlinico of Bari, University of Bari "Aldo Moro", Bari, Italy
| | - Antonella Bianco
- Department of Translational Biomedicine and Neurosciences (DiBraiN), Multiple Sclerosis Center - Policlinico of Bari, University of Bari "Aldo Moro", Bari, Italy
| | - Damiano Paolicelli
- Department of Translational Biomedicine and Neurosciences (DiBraiN), Multiple Sclerosis Center - Policlinico of Bari, University of Bari "Aldo Moro", Bari, Italy
| | - Pietro Iaffaldano
- Department of Translational Biomedicine and Neurosciences (DiBraiN), Multiple Sclerosis Center - Policlinico of Bari, University of Bari "Aldo Moro", Bari, Italy.
| |
Collapse
|
2
|
Siavoshi F, Ladakis DC, Muller A, Nourbakhsh B, Bhargava P. Ocrelizumab alters the circulating metabolome in people with relapsing-remitting multiple sclerosis. Ann Clin Transl Neurol 2024; 11:2485-2498. [PMID: 39185939 PMCID: PMC11537130 DOI: 10.1002/acn3.52167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Circulating metabolite levels are altered in multiple sclerosis (MS) and are associated with MS severity. However, how metabolic profiles shift following highly efficacious therapies, like ocrelizumab remains unclear. OBJECTIVE Circulating metabolite levels are altered in multiple sclerosis (MS) and are associated with MS severity. However, how metabolic profiles shift following highly efficacious therapies, like ocrelizumab remains unclear. To assess changes in the circulating metabolome produced by ocrelizumab treatment in people with relapsing-remitting MS (RRMS). METHODS Thirty-one individuals with RRMS eligible for beginning treatment with ocrelizumab were recruited and followed with demographic, clinical, quality-of-life, and global metabolomics data collected at each visit. Modules of highly correlated metabolites were identified using the weighted correlation network analysis approach. Changes in each module's eigenmetabolite values and individual metabolites during the study were evaluated using linear mixed-effects models. RESULTS Patients with a mean age of 40.8 (SD = 10.30) years, and median disease duration of 4.0 (IQR = 8.5) years, were monitored for a median of 3.36 (IQR = 1.43) years. Two out of twelve identified sets of metabolites were altered significantly. The first module mainly contained androgenic and pregnenolone steroids (p-value <0.001, coefficient: -0.10). The second module primarily consisted of several lysophospholipids, arachidonic acid, some endocannabinoids, and monohydroxy fatty acid metabolites (p-value = 0.016, coefficient: -0.12), which its reduction was significantly associated with improvement based on overall disability response score (OR 3.09e-01, 95% CI: 6.83e-02, 9.09e-01, p-value = 3.15E-02). INTERPRETATION In this longitudinal observational study, using a global untargeted metabolomics approach, we showed significant alteration in circulating metabolome in RRMS patients undergoing ocrelizumab treatment. In particular, we observed a significant reduction in metabolites involved in the lysophospholipid pathway, which was associated with patients' improvement.
Collapse
Affiliation(s)
- Fatemeh Siavoshi
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Dimitrios C. Ladakis
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Ashley Muller
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bardia Nourbakhsh
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Pavan Bhargava
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
3
|
Monteiro I, Nicolella V, Fiorenza M, Novarella F, Carotenuto A, Lanzillo R, Mauriello L, Scalia G, Castaldo G, Terracciano D, Brescia Morra V, Moccia M. The ocrelizumab wearing-off phenomenon is associated with reduced immunomodulatory response and increased neuroaxonal damage in multiple sclerosis. J Neurol 2024; 271:5012-5024. [PMID: 38777960 PMCID: PMC11319527 DOI: 10.1007/s00415-024-12434-w] [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: 02/15/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE The wearing-off phenomenon is common in people with multiple sclerosis (MS) treated with ocrelizumab. We aim to evaluate the presence and severity of wearing-off to ocrelizumab in relation to demographic and MS clinical variables, immune profiling, and a marker of neuroaxonal damage (plasma neurofilament light chain (pNfl)). METHODS This cross-sectional study included MS patients treated with ocrelizumab from at least 1 year. Wearing-off questionnaire and blood samples were collected between 21 and 23 weeks after the previous ocrelizumab infusion. Lymphocyte subpopulations were evaluated on peripheral blood using flow cytometry. PNfl was evaluated using fully automated chemiluminescent enzyme immunoassay. RESULTS We included 106 people with MS (age 49.5 ± 11.6 years; females 42.3%; wearing-off 57.6%). On regression models, wearing-off was associated with higher pNfl, CD8, CD3, and CD3CD27 lymphocytes. Most frequent wearing-off symptoms were cognitive, sensory, and balance problems; wearing-off started < 1 week (9.4%), 1-4 weeks (10.7%) or > 4 weeks (10.7%) before infusion; 44.8% of the complaints were moderate to severe. Severity of wearing-off was associated with higher pNfl and CD8 lymphocytes. CONCLUSIONS Wearing-off is common in people with MS treated with ocrelizumab, and is associated with reduced immunomodulation (higher T lymphocytes) and increased neuroaxonal damage, suggesting reduced treatment response.
Collapse
Affiliation(s)
- Isabel Monteiro
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
- Multiple Sclerosis Unit, Policlinico Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy
- Neurology Department, Coimbra University Hospital Center, Coimbra, Portugal
| | - Valerio Nicolella
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, Federico II University of Naples, Naples, Italy
| | - Mariano Fiorenza
- Department of Translational Medical Science, Federico II University of Naples, Naples, Italy
| | - Federica Novarella
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, Federico II University of Naples, Naples, Italy
| | - Antonio Carotenuto
- Multiple Sclerosis Unit, Policlinico Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, Federico II University of Naples, Naples, Italy
| | - Roberta Lanzillo
- Multiple Sclerosis Unit, Policlinico Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, Federico II University of Naples, Naples, Italy
| | | | - Giulia Scalia
- Centre for Advanced Biotechnology (CEINGE), Naples, Italy
| | - Giuseppe Castaldo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
- Centre for Advanced Biotechnology (CEINGE), Naples, Italy
| | - Daniela Terracciano
- Department of Translational Medical Science, Federico II University of Naples, Naples, Italy
| | - Vincenzo Brescia Morra
- Multiple Sclerosis Unit, Policlinico Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Neuroscience, Reproductive Sciences and Odontostomatology, Federico II University of Naples, Naples, Italy
| | - Marcello Moccia
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy.
- Multiple Sclerosis Unit, Policlinico Federico II University Hospital, Via Sergio Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
4
|
Björnsson GS, Sigurgrímsdóttir H, Maggadóttir SM, Einarsdóttir BÓ, Sveinsson ÓÁ, Hjaltason H, Sigurðardóttir SÞ, Lúðvíksson BR, Brynjólfsson SF. Long-term use of rituximab increases T cell count in MS patients. Front Immunol 2024; 15:1412668. [PMID: 39086478 PMCID: PMC11288840 DOI: 10.3389/fimmu.2024.1412668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Rituximab has been used to treat MS patients in Iceland for over a decade. However, long-term effect of rituximab on leukocyte populations has not yet been elucidated. By retrospective analysis of flow cytometric data from 349 patients visiting the neurological ward at The National University Hospital of Iceland from 2012 to 2023 for rituximab treatment, the long-term effect of rituximab and whether the effect was dose dependent (1000mg vs 500mg) was evaluated. No difference was detected in efficacy of B cell depletion in patients treated with 500mg as an initial dose of rituximab when compared to 1000mg. Long-term use of rituximab led to an increase in T cell count (p=0,0015) in patients receiving 3-8 doses of rituximab (1.5-8 years of treatment). The increase occurred in both CD4+ (p=0,0028) and CD8+ T cells (p=0,0015) and led to a decrease in the CD4/CD8 ratio (p=0,004). The most notable difference lies in reshaping the balance between näive and effector CD8+ T cells. The clinical implications of long-term treatment with rituximab and its effect on the T cell pool needs to be explored further. Since no difference in B cell depletion was detected between the two patient groups, 1000mg as an initial dose might be excessive, suggesting a personalized dosing regimen might have therapeutic and financial advantages.
Collapse
Affiliation(s)
| | - Hildur Sigurgrímsdóttir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Immunology, Landspítali – The National University Hospital of Iceland, Reykjavik, Iceland
| | - Sólrún Melkorka Maggadóttir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Immunology, Landspítali – The National University Hospital of Iceland, Reykjavik, Iceland
| | | | - Ólafur Árni Sveinsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Neurology, Landspítali – The National University Hospital of Iceland, Reykjavik, Iceland
| | - Haukur Hjaltason
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Neurology, Landspítali – The National University Hospital of Iceland, Reykjavik, Iceland
| | | | - Björn Rúnar Lúðvíksson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Immunology, Landspítali – The National University Hospital of Iceland, Reykjavik, Iceland
| | - Siggeir Fannar Brynjólfsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Immunology, Landspítali – The National University Hospital of Iceland, Reykjavik, Iceland
| |
Collapse
|
5
|
Langer-Gould A, Li BH, Smith JB, Xu S. Multiple Sclerosis, Rituximab, Hypogammaglobulinemia, and Risk of Infections. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200211. [PMID: 38507657 PMCID: PMC10959169 DOI: 10.1212/nxi.0000000000200211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/22/2023] [Indexed: 03/22/2024]
Abstract
BACKGROUND AND OBJECTIVES B-cell-depleting therapies increase the risk of infections and hypogammaglobulinemia. These relationships are poorly understood. The objectives of these analyses were to estimate how much of this rituximab-associated infection risk is mediated by hypogammaglobulinemia and to identify other modifiable risk factors in persons with multiple sclerosis (pwMS). METHODS We conducted a retrospective cohort study of rituximab-treated pwMS from January 1, 2008, to December 31, 2020, in Kaiser Permanente Southern California. Cumulative rituximab dose was defined as ≤2, >2 and ≤4, or >4 g. Serious infections were defined as infections requiring or prolonging hospitalizations, and recurrent outpatient infections as seeking care for ≥3 within 12 months. Exposures, outcomes, and covariates were collected from the electronic health record. Adjusted hazard ratios (aHRs) were estimated using Andersen-Gill hazards models, and generalized estimating equations were used to examine correlates of IgG values. Cross-sectional causal mediation analyses of rituximab and hypogammaglobulinemia were conducted. RESULTS We identified 2,482 pwMS who were treated with rituximab for a median of 2.4 years (interquartile range = 1.3-3.9). The average age at rituximab initiation was 43.0 years, 71.9% were female, 49.7% were White, non-Hispanic patients, and 29.6% had advanced disability (requiring walker or worse). Seven hundred patients (28.2%) developed recurrent outpatient infections, 155 (6.2%) developed serious infections, and only 248 (10.0%) had immunoglobulin G (IgG) < 700 mg/dL. Higher cumulative rituximab dose (>4 g) was correlated with lower IgG levels (Beta = -58.8, p < 0.0001, ref ≤2 g) and, in models mutually adjusted for hypogammaglobulinemia, both were independently associated with an increased risk of serious (>4 g, aHR = 1.56, 95% CI 1.09-2.24; IgG < 500, aHR = 2.98, 95% CI 1.56-5.72) and outpatient infections (>4 g, aHR = 1.73, 95% CI 1.44-2.06; IgG < 500 aHR = 2.06, 95% CI 1.52-2.80; ref = IgG ≥ 700). Hypogammaglobulinemia explained at most 17.9% (95% CI -47.2-119%) of serious infection risk associated with higher cumulative rituximab exposure but was not significant for outpatient infections. Other independent modifiable risk factors were advanced physical disability for serious (aHR = 5.51, 95% CI 3.71-8.18) and outpatient infections (aHR = 1.24, 95% CI 1.06-1.44) and COPD (aHR = 1.68, 95% CI 1.34-2.11) and obesity (aHR = 1.25, 95% CI 1.09-1.45) for outpatient infections. DISCUSSION Higher cumulative rituximab doses increase the risk of infections even in this population where 90% of patients maintained normal IgG levels. Clinicians should strive to use minimally effective doses of rituximab and other B-cell-depleting therapies and consider important comorbidities to minimize risks of infections.
Collapse
Affiliation(s)
- Annette Langer-Gould
- From the Department of Neurology (A.L.-G.), Los Angeles Medical Center, Southern California Permanente Medical Group; Departments of Clinical Science (A.L.-G.) and Health Systems Science (S.X.), Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena; Department of Research and Evaluation (B.H.L., J.B.S., S.X.), Southern California Permanente Medical Group, Pasadena, CA
| | - Bonnie H Li
- From the Department of Neurology (A.L.-G.), Los Angeles Medical Center, Southern California Permanente Medical Group; Departments of Clinical Science (A.L.-G.) and Health Systems Science (S.X.), Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena; Department of Research and Evaluation (B.H.L., J.B.S., S.X.), Southern California Permanente Medical Group, Pasadena, CA
| | - Jessica B Smith
- From the Department of Neurology (A.L.-G.), Los Angeles Medical Center, Southern California Permanente Medical Group; Departments of Clinical Science (A.L.-G.) and Health Systems Science (S.X.), Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena; Department of Research and Evaluation (B.H.L., J.B.S., S.X.), Southern California Permanente Medical Group, Pasadena, CA
| | - Stanley Xu
- From the Department of Neurology (A.L.-G.), Los Angeles Medical Center, Southern California Permanente Medical Group; Departments of Clinical Science (A.L.-G.) and Health Systems Science (S.X.), Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena; Department of Research and Evaluation (B.H.L., J.B.S., S.X.), Southern California Permanente Medical Group, Pasadena, CA
| |
Collapse
|
6
|
Elgenidy A, Abdelhalim NN, Al-kurdi MAM, Mohamed LA, Ghoneim MM, Fathy AW, Hassaan HK, Anan A, Alomari O. Hypogammaglobulinemia and infections in patients with multiple sclerosis treated with anti-CD20 treatments: a systematic review and meta-analysis of 19,139 multiple sclerosis patients. Front Neurol 2024; 15:1380654. [PMID: 38699050 PMCID: PMC11063306 DOI: 10.3389/fneur.2024.1380654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/03/2024] [Indexed: 05/05/2024] Open
Abstract
Background Recent years have seen the emergence of disease-modifying therapies in multiple sclerosis (MS), such as anti-cluster of differentiation 20 (anti-CD20) monoclonal antibodies, aiming to modulate the immune response and effectively manage MS. However, the relationship between anti-CD20 treatments and immunoglobulin G (IgG) levels, particularly the development of hypogammaglobulinemia and subsequent infection risks, remains a subject of scientific interest and variability. We aimed to investigate the intricate connection between anti-CD20 MS treatments, changes in IgG levels, and the associated risk of hypogammaglobulinemia and subsequent infections. Method PubMed, Scopus, Embase, Cochrane, and Web of Science databases have been searched for relevant studies. The "R" software utilized to analyze the occurrence of hypogammaglobulinemia, infections and mean differences in IgG levels pre- and post-treatment. The subgrouping analyses were done based on drug type and treatment duration. The assessment of heterogeneity utilized the I2 and chi-squared tests, applying the random effect model. Results Thirty-nine articles fulfilled our inclusion criteria and were included in our review which included a total of 20,501 MS patients. The overall prevalence rate of hypogammaglobulinemia was found to be 11% (95% CI: 0.08 to 0.15). Subgroup analysis based on drug type revealed varying prevalence rates, with rituximab showing the highest at 18%. Subgroup analysis based on drug usage duration revealed that the highest proportion of hypogammaglobulinemia occurred in individuals taking the drugs for 1 year or less (19%). The prevalence of infections in MS patients with a focus on different infection types stratified by the MS drug used revealed that pulmonary infections were the most prevalent (9%) followed by urinary tract infections (6%), gastrointestinal infections (2%), and skin and mucous membrane infections (2%). Additionally, a significant decrease in mean IgG levels after treatment compared to before treatment, with a mean difference of 0.57 (95% CI: 0.22 to 0.93). Conclusion This study provides a comprehensive analysis of the impact of anti-CD20 drugs on serum IgG levels in MS patients, exploring the prevalence of hypogammaglobulinemia, based on different drug types, treatment durations, and infection patterns. The identified rates and patterns offer a foundation for clinicians to consider in their risk-benefit. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=518239, CRD42024518239.
Collapse
Affiliation(s)
- Anas Elgenidy
- Faculty of Medicine, Cairo University, Giza, Egypt
- Karl-Jaspers-Klinik, Bad Zwischenahn, Germany
| | | | | | | | | | | | | | - Ahmed Anan
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Omar Alomari
- Hamidiye International School of Medicine, University of Health Sciences, Istanbul, Türkiye
| |
Collapse
|
7
|
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
|
8
|
Zettl UK, Rommer PS, Aktas O, Wagner T, Richter J, Oschmann P, Cepek L, Elias-Hamp B, Gehring K, Chan A, Hecker M. Interferon beta-1a sc at 25 years: a mainstay in the treatment of multiple sclerosis over the period of one generation. Expert Rev Clin Immunol 2023; 19:1343-1359. [PMID: 37694381 DOI: 10.1080/1744666x.2023.2248391] [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: 06/07/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Interferon beta (IFN beta) preparations are an established group of drugs used for immunomodulation in patients with multiple sclerosis (MS). Subcutaneously (sc) applied interferon beta-1a (IFN beta-1a sc) has been in continuous clinical use for 25 years as a disease-modifying treatment. AREAS COVERED Based on data published since 2018, we discuss recent insights from analyses of the pivotal trial PRISMS and its long-term extension as well as from newer randomized studies with IFN beta-1a sc as the reference treatment, the use of IFN beta-1a sc across the patient life span and as a bridging therapy, recent data regarding the mechanisms of action, and potential benefits of IFN beta-1a sc regarding vaccine responses. EXPERT OPINION IFN beta-1a sc paved the way to effective immunomodulatory treatment of MS, enabled meaningful insights into the disease process, and remains a valid therapeutic option in selected vulnerable MS patient groups.
Collapse
Affiliation(s)
- Uwe Klaus Zettl
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Paulus Stefan Rommer
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | | | | | | | | | | | - Andrew Chan
- Department of Neurology, Inselspital Bern, University Hospital Bern, Bern, Switzerland
| | - Michael Hecker
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
9
|
Ruggieri S, Aiello A, Tortorella C, Navarra A, Vanini V, Meschi S, Lapa D, Haggiag S, Prosperini L, Cuzzi G, Salmi A, Quartuccio ME, Altera AMG, Garbuglia AR, Ascoli Bartoli T, Galgani S, Notari S, Agrati C, Puro V, Nicastri E, Gasperini C, Goletti D. Dynamic Evolution of Humoral and T-Cell Specific Immune Response to COVID-19 mRNA Vaccine in Patients with Multiple Sclerosis Followed until the Booster Dose. Int J Mol Sci 2023; 24:ijms24108525. [PMID: 37239872 DOI: 10.3390/ijms24108525] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
This study characterizes antibody and T-cell immune responses over time until the booster dose of COronaVIrus Disease 2019 (COVID-19) vaccines in patients with multiple sclerosis (PwMS) undergoing different disease-modifying treatments (DMTs). We prospectively enrolled 134 PwMS and 99 health care workers (HCWs) having completed the two-dose schedule of a COVID-19 mRNA vaccine within the last 2-4 weeks (T0) and followed them 24 weeks after the first dose (T1) and 4-6 weeks after the booster (T2). PwMS presented a significant reduction in the seroconversion rate and anti-receptor-binding domain (RBD)-Immunoglobulin (IgG) titers from T0 to T1 (p < 0.0001) and a significant increase from T1 to T2 (p < 0.0001). The booster dose in PwMS showed a good improvement in the serologic response, even greater than HCWs, as it promoted a significant five-fold increase of anti-RBD-IgG titers compared with T0 (p < 0.0001). Similarly, the T-cell response showed a significant 1.5- and 3.8-fold increase in PwMS at T2 compared with T0 (p = 0.013) and T1 (p < 0.0001), respectively, without significant modulation in the number of responders. Regardless of the time elapsed since vaccination, most ocrelizumab- (77.3%) and fingolimod-treated patients (93.3%) showed only a T-cell-specific or humoral-specific response, respectively. The booster dose reinforces humoral- and cell-mediated-specific immune responses and highlights specific DMT-induced immune frailties, suggesting the need for specifically tailored strategies for immune-compromised patients to provide primary prophylaxis, early SARS-CoV-2 detection and the timely management of COVID-19 antiviral treatments.
Collapse
Affiliation(s)
- Serena Ruggieri
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Carla Tortorella
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Assunta Navarra
- Clinical Epidemiology Unit, National Institute for Infectious Disease Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Valentina Vanini
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
- UOS Professioni Sanitarie Tecniche, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Silvia Meschi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Daniele Lapa
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Shalom Haggiag
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Luca Prosperini
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Gilda Cuzzi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Andrea Salmi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | | | - Anna Maria Gerarda Altera
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Anna Rosa Garbuglia
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Tommaso Ascoli Bartoli
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Simonetta Galgani
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Stefania Notari
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Chiara Agrati
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, 00146 Rome, Italy
| | - Vincenzo Puro
- UOC Emerging Infections and Centro di Riferimento AIDS (CRAIDS), National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Emanuele Nicastri
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Claudio Gasperini
- Department of Neurosciences, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| |
Collapse
|